WO2009122180A1 - Pyrimidine derivatives capable of inhibiting one or more kinases - Google Patents

Pyrimidine derivatives capable of inhibiting one or more kinases Download PDF

Info

Publication number
WO2009122180A1
WO2009122180A1 PCT/GB2009/000878 GB2009000878W WO2009122180A1 WO 2009122180 A1 WO2009122180 A1 WO 2009122180A1 GB 2009000878 W GB2009000878 W GB 2009000878W WO 2009122180 A1 WO2009122180 A1 WO 2009122180A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
group
optionally substituted
compound
formula
Prior art date
Application number
PCT/GB2009/000878
Other languages
French (fr)
Inventor
Edward Giles Mciver
Justin Stephen Bryans
Ela Smiljanic
Stephen John Lewis
Joanne Hough
Thomas Drake
Original Assignee
Medical Research Council
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GB0806005A external-priority patent/GB0806005D0/en
Priority claimed from GB0812580A external-priority patent/GB0812580D0/en
Priority claimed from GB0822635A external-priority patent/GB0822635D0/en
Application filed by Medical Research Council filed Critical Medical Research Council
Publication of WO2009122180A1 publication Critical patent/WO2009122180A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/14Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to pyrimidine compounds that are capable of inhibiting one or more kinases.
  • the compounds find applications in the treatment of a variety of disorders, including cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, primary open angle glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and/or chronic inflammation.
  • Pyrimidines and analogues thereof are already described as active ingredients, such as, for example, the 2-anilino-pyrimidines as fungicides (DE-A-4029650) or substituted pyrimidine derivatives for treating neurological or neurodegenerative diseases (WO 99/19305).
  • pyrimidine derivatives As CDK inhibitors, the most varied pyrimidine derivatives are described, for example, bis(anilino)- pyrimidine derivatives (WO 00/12486), 2-amino-4-substituted pyrimidines (WO 01/14375), purines (WO 99/02162), 5-cyano-pyrimidines (WO 02/04429), anilinopyrimidines (WO 00/12486) and 2-N-dimethylaminopropoxy-pyrimidines (WO 00/39101).
  • bis(anilino)- pyrimidine derivatives WO 00/12486
  • 2-amino-4-substituted pyrimidines WO 01/14375
  • purines WO 99/02162
  • 5-cyano-pyrimidines WO 02/04429
  • anilinopyrimidines WO 00/12486
  • 2-N-dimethylaminopropoxy-pyrimidines WO 00/39101
  • a first aspect of the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt or ester thereof,
  • R 1 is C 3-8 -cycloalkyl
  • X is O, NR 7 or C ⁇ -heterocycloalkyl
  • R 2 is aryl, heteroaryl, fused or unfused aryl-C ⁇ -heterocycloalkyl or fused or unfused heteroaryl-C 3 .
  • 6-heterocycloalkyl each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci- 6 -alkyl, C 3 -7-cycloalkyl and a group A, wherein said group is optionally substituted by one or more substituents selected from aryl, heteroaryl, R 10 and a group A, said heteroaryl group is optionally substituted by one or more R 10 groups; and wherein said C 3-6 - heterocycloalkyl group optionally contains one or more groups selected from oxygen, sulfur, nitrogen and CO;
  • R 3 is Ci_ 6 -alkyl optionally substituted by one or more substituents selected from aryl, heteroaryl, -NR > 4 4 ⁇ R > 5 3 , - vN ⁇ R> 7XCO)NR 4 R', -NR 7 SO 2 R 6 , -NR 'COOR', -CONR >4 4 rR> 5 5 , C 3-6 -heterocycloalky] and
  • aryl, heteroaryl and C ⁇ -heterocycloalkyl groups are each optionally substituted by one or more substituents selected from -Ci. 6 -alkyl and a group A, wherein said -C ⁇ -alky!
  • A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -NO 2 , -NH 2 , -NR 4 R 5 , -OR 6 , -NR 7 (CO)R 6 , -NR 7 (CO)NR 4 R 5 , -NR 7 COOR 7 , -NR 7 (SO 2 )R 6 , - CO 2 H, -NR 7 (SO 2 )NR 4 R 5 , -COOR 7 , -CONR 4 R 5 , COR 6 , SO 2 NR 4 R 5 and -SO 2 R 6 ;
  • each R 4 and R 5 is independently selected from hydrogen, C 3-7 -cycloalkyl, aryl, heteroaryl, Ci- ⁇ -alkyl and a C 3 ⁇ -heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more R 10 groups, wherein said C ⁇ -alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR 8 R 9 , -NR 7 (CO)R 6 , NR 7 COOR 6 , -NR 7 (SO 2 )R 6 , -COOR 6 , -CONR 8 R 9 , OR 10 , -SO 2 R 6 and a C ⁇ -heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more or R 10 groups; or
  • R 4 and R 5 together with the N to which they are attached form a ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C ⁇ -heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR 8 R 9 and R 1D groups;
  • each R 6 is independently selected from C ⁇ -alkyl, C 3-7 cycloalkyl, C 4 . 7 -heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R 10 and -NR 8 R 9 ;
  • each R 7 is selected from hydrogen, is optionally substituted by one or more halogens;
  • each of R 8 and R 9 is independently selected from hydrogen and Ci_ 5 -alkyl, wherein said Ci ⁇ -alkyl group is optionally substituted by one or more halogens; or
  • R 8 and R 9 together with the N to which they are attached form a C 4- 6-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C ⁇ -heterocycloalkyl ring is optionally substituted by one or more R 10 groups; and each R 10 is selected from halogen, C 3 . 7 -cycloalkyl and C ]-6 -alkyl optionally substituted by one or more halogens, wherein where R 10 is C ⁇ -alkyl and two or more R 10 groups are attached to the same carbon atom, the R 10 groups may be linked to form a spiroalkyl group.
  • a second aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound as described above and a pharmaceutically acceptable carrier, diluent or excipient.
  • a third aspect of the invention relates to a compound as described above for use in medicine.
  • a fourth aspect of the invention relates to a compound as described above for use in treating a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • POAG Primary open Angle Glaucoma
  • a fifth aspect of the invention relates to the use of a compound as described above in the preparation of a medicament for treating or preventing a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, primary open angle glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, primary open angle glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, primary open angle glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis
  • a sixth aspect of the invention relates to the use of a compound as described above in the preparation of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity, wherein the kinase is selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2,
  • a seventh aspect of the invention relates to a method of treating a mammal having a disease state alleviated by the inhibition of a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound as described above.
  • a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon
  • An eighth aspect of the invention relates to the use of a compound as described above in an assay for identifying further candidate compounds capable of inhibiting one or more kinases selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon.
  • a ninth aspect of the invention relates to a process for preparing a compound of formula IV, wherein R 1 , R 2 , R 3 and R 7 are as defined above, said process comprising the steps of:
  • a tenth aspect of the invention relates to a process for preparing a compound of formula XIX, wherein R 1 , R 2 and R 3 are as defined above, said process comprising the steps of:
  • Alkyl is defined herein as a straight-chain or branched alkyl radical, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, hexyl.
  • the alkyl group is a C 1-6 alkyl group, more preferably a C M group.
  • Cycloalkyl is defined herein as a monocyclic alkyl ring, such as, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl.
  • the cycloalkyl group is a C3. 12 cycloalkyl group, more preferably a C 3-6 cycloalkyl group.
  • Halogen is defined herein as chloro, fluoro, bromo or iodo.
  • aryl refers to a C 6-12 aromatic group, which may be benzocondensed, for example, phenyl or naphthyl.
  • Heteroaryl is defined herein as a monocyclic or bicyclic C 2-12 aromatic ring comprising one or more heteroatoms (that may be the same or different), such as oxygen, nitrogen or sulfur. Examples of suitable heteroaryl groups include thienyl, furanyl, pyrrolyl, pyridinyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl etc.
  • Heterocycloalkyl refers to a cyclic aliphatic group containing one or more heteroatoms selected from nitrogen, oxygen and sulfur, which is optionally interrupted by one or more -(CO)- groups in the ring and/or which optionally contains one or more double bonds in the ring.
  • the heterocycloalkyl group comprises 3-6 carbon atoms and is fully saturated.
  • Preferred heterocycloalkyl groups include piperidinyl, pyrrolidinyl, piperazinyl, thiomorpholinyl and morpholinyl. More preferably, the heterocycloalkyl group is selected from N-piperidinyl, N- pyrrolidinyl, N-piperazinyl, N-thiomorpholinyl and N-morpholinyl.
  • R 1 is cyclopropyl or cyclobutyl.
  • R 1 is cyclopropyl.
  • X is O or NR 7 , more preferably, O, NMe or NH. Even more preferably, X is NH.
  • A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -NO 2 , -NH 2 , -NR 4 R 5 , -OR 6 , -NR 7 (CO)R 6 , -NR 7 (CO)NR 4 R 5 , -NR 7 COOR 7 , -NR 7 (SO 2 )R 6 , - CO 2 H, -NR 7 (SO 2 )NR 4 R 5 , -COOR 7 , -CONR 4 R 5 , COR 6 and -SO 2 CH 3 .
  • R 2 is an optionally substituted aryl or heteroaryl group selected from the following:
  • the substituent attached to the aryl or heteroaryl group is selected from C 3-7 - cycloalkyl and a group A, wherein said group is in turn optionally substituted by one or more substituents selected from aryl, heteroaryl, R 10 and a group A.
  • R 2 is an optionally substituted aryl or heteroaryl group selected from the following:
  • R 2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from C J-6 -alkyl and a group A, wherein said Ci -6 - alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A, and wherein A is selected from halogen, OH, CN, CF 3 , -NH 2 , -NR 4 R 5 , -OR 6 , NR 7 (CO)R 6 , -NR 7 COOR 7 , -NR 7 (SO 2 )R 6 , -COOH, -COOR 7 and CONR 4 R 5 .
  • R 2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from halogen, CN, NHCO-
  • R 2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from Me, Cl, F, CN, NHCOMe, CF 3 , COOH, CONH 2 , OH, NH 2 , NHSO 2 Me, OCF 3 , -NHCOO 1 Bu, -CO 2 Me, -NMe 2 , 4-methylpiperazin-l-yl, N-morpholinyl, (4- methylpiperazin- 1 -yl)-CO-, (N-morpholinyl)-CH 2 CH 2 O-, (imidazol- 1 -yl)-CH 2 - and
  • R 2 is a phenyl group optionally substituted by one or more groups selected from C ]-6 -alkyl, heteroaryl and A, wherein said and heteroaryl groups are in turn optionally substituted.
  • R 2 is a phenyl group substituted by one or more A groups.
  • the A group is selected from CF 3 , halogen, CN, NHSO 2 Me, SO 2 NR 4 R 5 , NR 4 R 5 , OR 6 , COOR 7 , NR 7 COOR 7 , NR 7 COR 6 , CONR 4 R 5 , NR 7 CONR 4 R 5 and NR 7 SO 2 R 6 .
  • R 2 is a phenyl group substituted by one or more groups, each of which in turn is optionally substituted by one or more groups selected from heteroaryl and A.
  • the heteroaryl group is selected from imidazolyl and triazolyl.
  • the A group is selected from CONR 4 R 5 , NR 4 R 5 , OR 6 , COOR 7 and CN.
  • R 2 is a phenyl group substituted by one or more heteroaryl groups.
  • the heteroaryl group is selected from pyrimidinyl, tetrazolyl, pyridinyl, pyrazolyl, oxazolyl and triazolyl.
  • R 2 is a pyridyl group optionally substituted by one or more groups selected from heteroaryl and A, wherein said and heteroaryl groups are in turn optionally substituted.
  • R 2 is a pyridyl group substituted by one or more A groups.
  • the A group is selected from NR 4 R 5 , halo and OR 6 .
  • R 2 is a pyridyl group substituted by a heteroaryl group.
  • the heteroaryl group is selected from pyrazolyl, pyrimidinyl and pyridinyl.
  • R 2 is a pyridyl group substituted by a Ci- ⁇ -alkyl group, wherein said Ci- 6 -alkyl group is in turn optionally substituted with one or more substituents selected from NR 4 R 5 and OR 6 .
  • R 2 is an optionally substituted fused aryl-Cs ⁇ -heterocycloalkyl or fused heteroaryl-C 3 ⁇ -heterocycloalkyl.
  • R 2 is selected from the following:
  • the substituent is an optionally substituted group.
  • R 3 is C ⁇ -alkyl optionally substituted by one or more substituents selected from heteroaryl, -NR 4 R 5 , -NR 7 (CO)R 6 , -NR 7 COOR 7 , C 3 ⁇ - heterocycloalkyl and
  • R 3 is Ci- 4 -alkyl substituted by -NR 7 (CO)R 6 .
  • R 7 is H and R 6 is selected from C ⁇ -alkyl, C 3 .7 cycloalkyl, C 4-7 -heterocycloalkyl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R 10 and -NR 8 R 9 .
  • R 3 is C M -alkyl substituted by -NR 7 (CO)R 6 and R 6 is selected from thienyl, cyclopentyl, CH 2 -cyclopentyl, methyl, isopropyl, n-propyl, pyrazolyl, cyclohexyl, thiazolyl, oxazolyl, furanyl, imidazolyl, cyclopropyl, CH 2 -cyclopropyl, cyclobutyl, triazolyl, pyrrolyl, tetrahydrofuranyl and isoxazolyl, each of which may be substituted by one or more R 10 or -NR 8 R 9 groups.
  • R 3 is C M -alkyl substituted by -NR 7 (CO)R 6 and R 6 is selected from thienyl, cyclopentyl, CH 2 -cyclopentyl, methyl, isopropyl, n-propyl, pyrazolyl, cyclohexyl, thiazolyl, oxazolyl, furanyl, imidazolyl, cyclopropyl, CH 2 -cyclopropyl, cyclobutyl, triazolyl, pyrrolyl, tetrahydrofuranyl and isoxazolyl, each of which may be substituted by one or more halogen or Ci -6 - alkyl groups.
  • R 3 is substituted by -NR 7 (CO)R 6 and R 6 is selected from thienyl, cyclopentyl, CH 2 -cyclopentyl, methyl, isopropyl, n-propyl, pyrazolyl, cyclohexyl, thiazolyl, oxazolyl, furanyl, CF 3 , imidazolyl, cyclopropyl, CH 2 -cyclopropyl, cyclobutyl, triazolyl, pyrrolyl, tetrahydrofuranyl, CH 2 NMe 2 and isoxazolyl.
  • R 3 is substituted by a heteroaryl group.
  • the heteroaryl group is selected from pyrazolyl, tetrazolyl and triazolyl.
  • R 3 is C M -alkyl substituted by a -NR 4 R 5 group.
  • R 4 and R 5 together with the N to which they are attached form a C ⁇ -heterocycloalkyl ring optionally further containing one or more CO groups, wherein said C ⁇ -heterocycloalkyl ring is optionally substituted with one or more R 10 groups.
  • R 10 is Ci -5 - alkyl optionally substituted with one or more halo atoms.
  • R 3 is C M -alkyl substituted by one of the following groups:
  • R 3 is Ci ⁇ -alkyl substituted by one of the following groups:
  • R 3 is C M -alkyl substituted by a Cs- ⁇ -heterocycloalkyl group, wherein said Cs- ⁇ -heterocycloalkyl group is optionally substituted by one or more A groups.
  • the A group is a COR 6 group.
  • R 3 is C ⁇ -alkyl substituted by a group selected from:
  • R is an optionally substituted C 3 -alkyl group.
  • the compound of the invention is selected from the following: 9 10 11 12 13 14 15 16
  • Example 194 Example 202
  • the compound of the invention is selected from compounds 1-214 above.
  • a further aspect of the invention relates to a compound as described above for use in medicine.
  • Another aspect of the invention relates to a compound as described above for use in treating a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • POAG Primary open Angle Glaucoma
  • Another aspect relates to the use of a compound as described above in the preparation of a medicament for treating or preventing a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
  • POAG Primary open Angle Glaucoma
  • the compound is administered in an amount sufficient to inhibit a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR and IKKepsilon.
  • a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR and IKKepsilon.
  • Yet another aspect relates to the use of a compound of the invention in the preparation of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity, wherein the kinase is selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, IKKepsilon and combinations thereof.
  • the kinase is selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, IKKepsilon and combinations thereof.
  • the kinase is selected from TBKl, PDKl, ERK8, MARK3, and IKKepsilon and combinations thereof.
  • the kinase is selected from TBKl and PDKl. Even more preferably, the kinase is TBKl .
  • the compound of the invention is exhibits an IC 50 value against TBKl of from about 1 ⁇ M to about 10 ⁇ M, more preferably from about 100 nM to about 1 ⁇ M, even more preferably, less than about 100 nM.
  • the disorder is selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, diseases of the eye, including Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, autoimmune diseases, artherosclerosis, retinopathy, osteoarthritis, fibrotic diseases, endometriosis and chronic inflammation.
  • POAG Primary open Angle Glaucoma
  • TLR4 Toll-like receptor 4
  • RNA from double stranded RNA viruses
  • TLR3 Toll-like receptor 4
  • IRF3 interferon-regulatory factor 3
  • IL-8 interleukin-8
  • RANTES interleukin-8
  • TBKl is also activated in response to hypoxia and stimulates the production of pro-angiogenic factors, such as VEGF and IL-I.
  • the expression of TBKl rises 2.5-3-fold after 24h of hypoxia, similar to the increase in expression of VEGF.
  • the hypoxia induced increase in VEGF expression can be abolished by siRNA "knockdown" of TBKl.
  • the level of TBKl mRNA and protein is elevated in malignant colon and breast cancer cells (see Korherr et al (2006) PNAS 103, 4240-4245 and references therein).
  • TBKl is also recruited and activated by the RalB/Sec5 effector complex; in cancer cells, constitutive engagement of this pathway via chronic RaIB activation, restricts the initiation of apoptotic programmes (Chien et al (2006) Cell 127, 157-170 and references there-in).
  • the drugs that inhibit TBKl may have efficacy for the treatment of cancers.
  • the compounds of the invention are useful in the treatment of Primary open Angle Glaucoma (POAG).
  • POAG Primary Open Angle Glaucoma
  • OPTN protein optineurin
  • NVG Normal Tension Glaucoma
  • LPG Low Tension Glaucoma
  • tumour necrosis factor ⁇ (TNF ⁇ ) has been reported to increase the severity of damage in optic nerve heads of POAG and LTG subjects 3 ' 4 .
  • exposure to TNF ⁇ 10 induces the de novo expression of optineurin.
  • Another aspect of the invention relates to a method of treating a TBKl, MKKl, ERK8, RSKl, RSK2, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and/or IKKepsilon and optionally PDKl related disease or disorder.
  • the method according to this aspect of the present invention is effected by administering to a subject in need thereof a therapeutically effective amount of a compound of the present invention, as described hereinabove, either per se, or, more preferably, as a part of a pharmaceutical composition, mixed with, for example, a pharmaceutically acceptable carrier, as is detailed hereinafter.
  • Yet another aspect of the invention relates to a method of treating a mammal having a disease state alleviated by the inhibition of a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound according to the invention.
  • a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon
  • the disease state is alleviated by the inhibition of TBKl, PDKl, ERK8, MARK3 or IKKepsilon, more preferably TBKl or IKKepsilon, even more preferably TBKl.
  • the mammal is a human.
  • method refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
  • administering refers to a method for bringing a compound of the present invention and a target kinase together in such a manner that the compound can affect the enzyme activity of the kinase either directly; i.e., by interacting with the kinase itself or indirectly; i.e., by interacting with another molecule on which the catalytic activity of the kinase is dependent.
  • administration can be accomplished either in vitro, i.e. in a test tube, or in vivo, i.e., in cells or tissues of a living organism.
  • treating includes abrogating, substantially inhibiting, slowing or reversing the progression of a disease or disorder, substantially ameliorating clinical symptoms of a disease or disorder or substantially preventing the appearance of clinical symptoms of a disease or disorder.
  • preventing refers to a method for barring an organism from acquiring a disorder or disease in the first place.
  • terapéuticaally effective amount refers to that amount of the compound being administered which will relieve to some extent one or more of the symptoms of the disease or disorder being treated.
  • a therapeutically effective amount can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC 50 Or the ICioo as determined in cell culture. Such information can be used to more accurately determine useful doses in humans.
  • Initial dosages can also be estimated from in vivo data. Using these initial guidelines one of ordinary skill in the art could determine an effective dosage in humans.
  • toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD 50 and the ED 50 .
  • the dose ratio between toxic and therapeutic effect is the therapeutic index and can be expressed as the ratio between LD 50 and ED 50 .
  • Compounds which exhibit high therapeutic indices are preferred.
  • the data obtained from these cell cultures assays and animal studies can be used in formulating a dosage range that is not toxic for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition, (see, e.g., Fingl et al., 1975, In: The Pharmacological Basis of Therapeutics, chapter 1, page 1).
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active compound which are sufficient to maintain therapeutic effect.
  • Usual patient dosages for oral administration range from about 50-2000 mg/kg/day, commonly from about 100-1000 mg/kg/day, preferably from about 150-700 mg/kg/day and most preferably from about 250-500 mg/kg/day.
  • therapeutically effective serum levels will be achieved by administering multiple doses each day.
  • the effective local concentration of the drug may not be related to plasma concentration.
  • One skilled in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
  • kinase related disease or disorder refers to a disease or disorder characterized by inappropriate kinase activity or over-activity of a kinase as defined herein. Inappropriate activity refers to either; (i) kinase expression in cells which normally do not express said kinase; (ii) increased kinase expression leading to unwanted cell proliferation, differentiation and/or growth; or, (iii) decreased kinase expression leading to unwanted reductions in cell proliferation, differentiation and/or growth.
  • Over-activity of kinase refers to either amplification of the gene encoding a particular kinase or production of a level of kinase activity, which can correlate with a cell proliferation, differentiation and/or growth disorder (that is, as the level of the kinase increases, the severity of one or more of the symptoms of the cellular disorder increases).
  • Over activity can also be the result of ligand independent or constitutive activation as a result of mutations such as deletions of a fragment of a kinase responsible for ligand binding.
  • Preferred diseases or disorders that the compounds described herein may be useful in preventing, treating and/or studying are cell proliferative disorders, especially cancer such as, but not limited to, papilloma, blastoglioma, Kaposi's sarcoma, melanoma, lung cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, astrocytoma, head cancer, neck cancer, skin cancer, liver cancer, bladder cancer, breast cancer, lung cancer, uterus cancer, prostate cancer, testis carcinoma, colorectal cancer, thyroid cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma, leukemia, lymphoma, Hodgkin's disease and Burkitt's disease.
  • cancer such as, but not limited to, papilloma, blastoglioma, Kaposi's sarcoma, melanoma, lung cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, astrocytoma, head cancer, neck cancer, skin cancer
  • Another condition to which the compounds described herein may be useful in preventing, treating and/or studying is septic shock.
  • Another condition to which the compounds described herein may be useful in preventing, treating and/or studying is inflammatory disease.
  • a further aspect relates to the use of a compound which is capable of inhibiting the binding of TBKl to a mutant form of OPTN for the manufacture of a medicament for treating POAG and/or a disease where it would be desirable to inhibit or reduce TBKl binding to mutant form of OPTN.
  • a mutant form of OPTN is the manufacture of a medicament for treating POAG and/or a disease where it would be desirable to inhibit or reduce TBKl binding to mutant form of OPTN.
  • One such mutant is the OPTN (E50K) mutant.
  • Suitable compounds may include the compounds identified herein.
  • a method of treating a patient suffering from POAG comprising the step of administering to the subject an effective amount of a compound which is capable of inhibiting an interaction between TBKl and a mutant form of OPTN, associated with POAG.
  • Suitable compounds include those according to Formula I.
  • a method of treating a patient suffering from a disease associated with abnormal cell proliferation comprising the step of administering to the subject an effective amount of a compound of the invention.
  • a method of treating a patient suffering from septic shock, neurodegenerative diseases, Alzheimer's disease comprising the step of administering to the subject an effective amount of a compound of the invention.
  • the present invention further provides use of compounds as defined herein for the manufacture of medicaments for the treatment of diseases where it is desirable to inhibit TBKl and/or IKK epsilon.
  • diseases include colon and breast cancer, septic shock and/or POAG.
  • a number of papers 5 ' 6 ' 7 have described that TBKl and IKKepsilon modulate expression of interferon and interferon inducible genes, without affecting induction of pro-inflammatory cytokines. This indicates that the compounds disclosed herein, may find applications in treating/preventing septic shock or viral infection. Mice that do not express interferon beta or IRF3 are resistant to lipopolysaccharide induced septic shock so that inhibitors of TBKl should be expected to have a similar effect.
  • the compounds or physiologically acceptable salt, ester or other physiologically functional derivative thereof, described herein may be presented as a pharmaceutical formulation, comprising the compounds or physiologically acceptable salt, ester or other physiologically functional derivative thereof, together with one or more pharmaceutically acceptable carriers therefore and optionally other therapeutic and/or prophylactic ingredients.
  • the carrier(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • the pharmaceutical compositions may be for human or animal usage in human and veterinary medicine.
  • suitable carriers include lactos ' e, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like.
  • suitable diluents include ethanol, glycerol and water.
  • compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s), buffer(s), flavouring agent(s), surface active agent(s), thickener(s), preservative(s) (including anti-oxidants) and the like, and substances included for the purpose of rendering the formulation isotonic with the blood of the intended recipient.
  • suitable binder(s) lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s), buffer(s), flavouring agent(s), surface active agent(s), thickener(s), preservative(s) (including anti-oxidants) and the like, and substances included for the purpose of rendering the formulation isotonic with the blood of the intended recipient.
  • Suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.
  • Suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • Antioxidants and suspending agents may be also used.
  • compositions include those suitable for oral, topical (including dermal, buccal and sublingual), rectal or parenteral (including subcutaneous, intradermal, intramuscular and intravenous), nasal and pulmonary administration e.g., by inhalation.
  • the formulation may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association an active compound with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • compositions suitable for oral administration wherein the carrier is a solid are most preferably presented as unit dose formulations such as boluses, capsules or tablets each containing a predetermined amount of active compound.
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine an active compound in a free-flowing form such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, lubricating agent, surface-active agent or dispersing agent.
  • Moulded tablets may be made by moulding an active compound with an inert liquid diluent. Tablets may be optionally coated and, if uncoated, may optionally be scored.
  • Capsules may be prepared by filling an active compound, either alone or in admixture with one or more accessory ingredients, into the capsule shells and then sealing them in the usual manner.
  • Cachets are analogous to capsules wherein an active compound together with any accessory ingredient(s) is sealed in a rice paper envelope.
  • An active compound may also be formulated as dispersible granules, which may for example be suspended in water before administration, or sprinkled on food. The granules may be packaged, e.g., in a sachet.
  • Formulations suitable for oral administration wherein the carrier is a liquid may be presented as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water liquid emulsion.
  • Formulations for oral administration include controlled release dosage forms, e.g., tablets wherein an active compound is formulated in an appropriate release - controlling matrix, or is coated with a suitable release - controlling film. Such formulations may be particularly convenient for prophylactic use.
  • compositions suitable for rectal administration wherein the carrier is a solid are most preferably presented as unit dose suppositories.
  • Suitable carriers include cocoa butter and other materials commonly used in the art.
  • the suppositories may be conveniently formed by admixture of an active compound with the softened or melted carriers) followed by chilling and shaping in moulds.
  • Pharmaceutical formulations suitable for parenteral administration include sterile solutions or suspensions of an active compound in aqueous or oleaginous vehicles.
  • Injectible preparations may be adapted for bolus injection or continuous infusion. Such preparations are conveniently presented in unit dose or multi-dose containers which are sealed after introduction of the formulation until required for use.
  • an active compound may be in powder form which is constituted with a suitable vehicle, such as sterile, pyrogen-free water, before use.
  • An active compound may also be formulated as long-acting depot preparations, which may be administered by intramuscular injection or by implantation, e.g., subcutaneously or intramuscularly. Depot preparations may include, for example, suitable polymeric or hydrophobic materials, or ion- exchange resins. Such long-acting formulations are particularly convenient for prophylactic use. Formulations suitable for pulmonary administration via the buccal cavity are presented such that particles containing an active compound and desirably having a diameter in the range of 0.5 to 7 microns are delivered in the bronchial tree of the recipient.
  • such formulations are in the form of finely comminuted powders which may conveniently be presented either in a pierceable capsule, suitably of, for example, gelatin, for use in an inhalation device, or alternatively as a self-propelling formulation comprising an active compound, a suitable liquid or gaseous propellant and optionally other ingredients such as a surfactant and/or a solid diluent.
  • suitable liquid propellants include propane and the chlorofluorocarbons
  • suitable gaseous propellants include carbon dioxide.
  • Self-propelling formulations may also be employed wherein an active compound is dispensed in the form of droplets of solution or suspension.
  • Such self-propelling formulations are analogous to those known in the art and may be prepared by established procedures. Suitably they are presented in a container provided with either a manually- operable or automatically functioning valve having the desired spray characteristics; advantageously the valve is of a metered type delivering a fixed volume, for example, 25 to 100 microlitres, upon each operation thereof.
  • a manually- operable or automatically functioning valve having the desired spray characteristics; advantageously the valve is of a metered type delivering a fixed volume, for example, 25 to 100 microlitres, upon each operation thereof.
  • an active compound may be in the form of a solution or suspension for use in an atomizer or nebuliser whereby an accelerated airstream or ultrasonic agitation is employed to produce a fine droplet mist for inhalation.
  • Formulations suitable for nasal administration include preparations generally similar to those described above for pulmonary administration. When dispensed such formulations should desirably have a particle diameter in the range 10 to 200 microns to enable retention in the nasal cavity; this may be achieved by, as appropriate, use of a powder of a suitable particle size or choice of an appropriate valve. Other suitable formulations include coarse powders having a particle diameter in the range 20 to 500 microns, for administration by rapid inhalation through the nasal passage from a container held close up to the nose, and nasal drops comprising 0.2 to 5% w/v of an active compound in aqueous or oily solution or suspension.
  • Pharmaceutically acceptable carriers are well known to those skilled in the art and include, but are not limited to, 0.1 M and preferably 0.05 M phosphate buffer or 0.8% saline. Additionally, such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, chelating agents, inert gases and the like.
  • Formulations suitable for topical formulation may be provided for example as gels, creams or ointments. Such preparations may be applied e.g. to a wound or ulcer either directly spread upon the surface of the wound or ulcer or carried on a suitable support such as a bandage, gauze, mesh or the like which may be applied to and over the area to be treated.
  • a suitable support such as a bandage, gauze, mesh or the like which may be applied to and over the area to be treated.
  • Liquid or powder formulations may also be provided which can be sprayed or sprinkled directly onto the site to be treated, e.g. a wound or ulcer.
  • a carrier such as a bandage, gauze, mesh or the like can be sprayed or sprinkle with the formulation and then applied to the site to be treated.
  • a process for the preparation of a pharmaceutical or veterinary composition as described above comprising bringing the active compound(s) into association with the carrier, for example by admixture.
  • the formulations are prepared by uniformly and intimately bringing into association the active agent with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • the invention extends to methods for preparing a pharmaceutical composition comprising bringing a compound of general formula (I) in conjunction or association with a pharmaceutically or veterinarily acceptable carrier or vehicle.
  • the compounds of the invention can be present as salts or esters, in particular pharmaceutically and veterinarily acceptable salts or esters.
  • salts of the compounds of the invention include suitable acid addition or base salts thereof.
  • suitable pharmaceutical salts may be found in Berge et al, J Pharm Sci, 66, 1-19 (1977). Salts are formed, for example with strong inorganic acids such as mineral acids, e.g.
  • hydrohalic acids such as hydrochloride, hydrobromide and hydroiodide, sulfuric acid, phosphoric acid sulphate, bisulphate, hemisulphate, thiocyanate, persulphate and sulphonic acids; with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Ci-C 4 )-alkyl- or aryl-sulfonic acids which
  • Preferred salts include, for example, acetate, trifluoroacetate, lactate, gluconate, citrate, tartrate, maleate, malate, pantothenate, adipate, alginate, aspartate, benzoate, butyrate, digluconate, cyclopentanate, glucoheptanate, glycerophosphate, oxalate, heptanoate, hexanoate, fumarate, nicotinate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, proprionate, tartrate, lactobionate, pivolate, camphorate, undecanoate and succinate, organic sulphonic acids such as methanesulphonate, ethanesulphonate, 2-hydroxyethane sulphonate, camphorsulphonate, 2- naphthalenesulphonate, benzenesulphonate, p-chlorobenzenesulphon
  • Esters are formed either using organic acids or alcohols/hydroxides, depending on the functional group being esterified.
  • Organic acids include carboxylic acids, such as alkanecarboxylic acids of 1 to 12 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acid, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Ci-C 4 )-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-tol
  • Suitable hydroxides include inorganic hydroxides, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminium hydroxide.
  • Alcohols include alkanealcohols of 1-12 carbon atoms which may be unsubstituted or substituted, e.g. by a halogen).
  • the invention includes, where appropriate all enantiomers, diastereoisomers and tautomers of the compounds of the invention.
  • the person skilled in the art will recognise compounds that possess optical properties (one or more chiral carbon atoms) or tautomeric characteristics.
  • the corresponding enantiomers and/or tautomers may be isolated/prepared by methods known in the art.
  • Enantiomers are characterised by the absolute configuration of their chiral centres and described by the R- and S-sequencing rules of Cahn, Ingold and Prelog. Such conventions are well known in the art (e.g. see 'Advanced Organic Chemistry', 3 rd edition, ed.
  • Compounds of the invention containing a chiral centre may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well-known techniques and an individual enantiomer may be used alone.
  • Some of the compounds of the invention may exist as stereoisomers and/or geometric isomers - e.g. they may possess one or more asymmetric and/or geometric centres and so may exist in two or more stereoisomeric and/or geometric forms.
  • the present invention contemplates the use of all the individual stereoisomers and geometric isomers of those inhibitor agents, and mixtures thereof.
  • the terms used in the claims encompass these forms, provided said forms retain the appropriate functional activity (though not necessarily to the same degree).
  • the present invention also includes all suitable isotopic variations of the agent or a pharmaceutically acceptable salt thereof.
  • An isotopic variation of an agent of the present invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature.
  • isotopes that can be incorporated into the agent and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine such as 2 H, 3 H, 13 C, 14 C, 15 N, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F and 36 Cl, respectively.
  • isotopic variations of the agent and pharmaceutically acceptable salts thereof are useful in drug and/or substrate tissue distribution studies. Tritiated, i.e., 3 H, and carbon- 14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium, i.e., 2 H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances.
  • the invention includes compounds of general formula (I) where any hydrogen atom has been replaced by a deuterium atom. Isotopic variations of the agent of the present invention and pharmaceutically acceptable salts thereof of this invention can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
  • PRODRUGS The invention further includes the compounds of the present invention in prodrug form, i.e. covalently bonded compounds which release the active parent drug according to general formula (I) in vivo.
  • prodrugs are generally compounds of the invention wherein one or more appropriate groups have been modified such that the modification may be reversed upon administration to a human or mammalian subject. Reversion is usually performed by an enzyme naturally present in such subject, though it is possible for a second agent to be administered together with such a prodrug in order to perform the reversion in vivo. Examples of such modifications include ester (for example, any of those described above), wherein the reversion may be carried out be an esterase etc. Other such systems will be well known to those skilled in the art.
  • the present invention also includes solvate forms of the compounds of the present invention.
  • the terms used in the claims encompass these forms.
  • the invention further relates to the compounds of the present invention in their various crystalline forms, polymorphic forms and (an)hydrous forms. It is well established within the pharmaceutical industry that chemical compounds may be isolated in any of such forms by slightly varying the method of purification and or isolation form the solvents used in the synthetic preparation of such compounds.
  • compositions of the present invention may be adapted for rectal, nasal, intrabronchial, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intraarterial and intradermal), intraperitoneal or intrathecal administration.
  • the formulation is an orally administered formulation.
  • the formulations may conveniently be presented in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
  • the formulations may be in the form of tablets and sustained release capsules, and may be prepared by any method well known in the art of pharmacy.
  • Formulations for oral administration in the present invention may be presented as: discrete units such as capsules, gellules, drops, cachets, pills or tablets each containing a predetermined amount of the active agent; as a powder or granules; as a solution, emulsion or a suspension of the active agent in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water- in-oil liquid emulsion; or as a bolus etc.
  • these compositions contain from 1 to 250 mg and more preferably from 10-100 mg, of active ingredient per dose.
  • the term "acceptable carrier” includes vehicles such as common excipients e.g. binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone (Povidone), methylcellulose, ethylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, sucrose and starch; fillers and carriers, for example corn starch, gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid; and lubricants such as magnesium stearate, sodium stearate and other metallic stearates, glycerol stearate stearic acid, silicone fluid, talc waxes, oils and colloidal silica.
  • Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring and the like can also be used. It may be desirable to
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active agent in a free flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent.
  • Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may be optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active agent.
  • compositions suitable for oral administration include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active agent in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active agent in a suitable liquid carrier.
  • Other forms of administration comprise solutions or emulsions which may be injected intravenously, intraarterially, intrathecally, subcutaneously, intradermally, intraperitoneally or intramuscularly, and which are prepared from sterile or sterilisable solutions. Injectable forms typically contain between 10 - 1000 mg, preferably between 10 - 250 mg, of active ingredient per dose.
  • compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders.
  • An alternative means of transdermal administration is by use of a skin patch.
  • the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin.
  • the active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
  • a person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation.
  • a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy.
  • the dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • an effective amount of a compound of general formula (I) may be administered to inhibit the kinase implicated with a particular condition or disease.
  • this dosage amount will further be modified according to the type of administration of the compound.
  • parenteral administration of a compound of general formula (I) is preferred.
  • An intravenous infusion of the compound in 5% dextrose in water or normal saline, or a similar formulation with suitable excipients, is most effective, although an intramuscular bolus injection is also useful.
  • the parenteral dose will be about 0.01 to about 100 mg/kg; preferably between 0.1 and 20 mg/kg, in a manner to maintain the concentration of drug in the plasma at a concentration effective to inhibit a kinase.
  • the compounds may be administered one to four times daily at a level to achieve a total daily dose of about 0.4 to about 400 mg/kg/day.
  • the precise amount of an inventive compound which is therapeutically effective, and the route by which such compound is best administered, is readily determined by one of ordinary skill in the art by comparing the blood level of the agent to the concentration required to have a therapeutic effect.
  • the compounds of this invention may also be administered orally to the patient, in a manner such that the concentration of drug is sufficient to achieve one or more of the therapeutic indications disclosed herein.
  • a pharmaceutical composition containing the compound is administered at an oral dose of between about 0.1 to about 50 mg/kg in a manner consistent with the condition of the patient.
  • the oral dose would be about 0.5 to about 20 mg/kg.
  • the compounds of this invention which may have good bioavailability, may be tested in one of several biological assays to determine the concentration of a compound which is required to have a given pharmacological effect.
  • the one or more compounds of the invention are administered in combination with one or more other active agents, for example, existing drugs available on the market.
  • the compounds of the invention may be administered consecutively, simultaneously or sequentially with the one or more other active agents.
  • Drugs in general are more effective when used in combination.
  • combination therapy is desirable in order to avoid an overlap of major toxicities, mechanism of action and resistance mechanism(s).
  • the major advantages of combining chemotherapeutic drugs are that it may promote additive or possible synergistic effects through biochemical interactions and also may decrease the emergence of resistance.
  • Beneficial combinations may be suggested by studying the inhibitory activity of the test compounds with agents known or suspected of being valuable in the treatment of a particular disorder. This procedure can also be used to determine the order of administration of the agents, i.e. before, simultaneously, or after delivery. Such scheduling may be a feature of all the active agents identified herein.
  • a further aspect of the invention relates to the use of a compound as described above in an assay for identifying further candidate compounds capable of inhibiting one or more kinases selected from TBK 1 , MKK 1 , ERO, RSK 1 , RSK2, PDK 1 , S6K 1 , MNK2, PHK, CHK 1 , CHK2, GSObeta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon.
  • the assay is a competitive binding assay.
  • the competitive binding assay comprises contacting a compound of the invention with a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, and a candidate compound and detecting any change in the interaction between the compound according to the invention and the kinase.
  • a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon
  • the candidate compound is generated by conventional SAR modification of a compound of the invention.
  • conventional SAR modification refers to standard methods known in the art for varying a given compound by way of chemical derivatisation.
  • the identified compound may act as a model (for example, a template) for the development of other compounds.
  • the compounds employed in such a test may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The abolition of activity or the formation of binding complexes between the compound and the agent being tested may be measured.
  • the assay of the present invention may be a screen, whereby a number of agents are tested.
  • the assay method of the present invention is a high through-put screen.
  • This invention also contemplates the use of competitive drug screening assays in which neutralising antibodies capable of binding a compound specifically compete with a test compound for binding to a compound.
  • HTS high throughput screening
  • the competitive binding assay comprises contacting a compound of the invention with a kinase in the presence of a known substrate of said kinase and detecting any change in the interaction between said kinase and said known substrate.
  • a further aspect of the invention provides a method of detecting the binding of a ligand to a kinase, said method comprising the steps of:
  • One aspect of the invention relates to a process comprising the steps of:
  • Another aspect of the invention provides a process comprising the steps of:
  • the invention also relates to a ligand identified by the method described hereinabove.
  • Yet another aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a ligand identified by the method described hereinabove.
  • Another aspect of the invention relates to the use of a ligand identified by the method described hereinabove in the preparation of a pharmaceutical composition for use in the treatment of one or more disorders selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, diseases of the eye, including Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, autoimmune diseases, artherosclerosis, retinopathy, osteoarthritis, f ⁇ brotic diseases, endometriosis and chronic inflammation.
  • POAG Primary open Angle Glaucoma
  • the above methods may be used to screen for a ligand useful as an inhibitor of one or more kinases.
  • a further aspect of the invention relates to a process for preparing a compound as described above, said process comprising the steps of:
  • Scheme 1 illustrates the conversion of compounds of formula (Ia) to compounds of formula (II), wherein Rl, R3 and R7 are as defined previously.
  • Compounds of formula (Ia) can be converted to compounds of formula (II) by treatment with POCl 3 in the presence of an additive, such as N,N-diisopropylethylamine at temperatures in the range of 5O 0 C to reflux for reaction times up to 24 h.
  • an additive such as N,N-diisopropylethylamine at temperatures in the range of 5O 0 C to reflux for reaction times up to 24 h.
  • Preferred conditions leq. of formula 1, leq of N,N-diisopropylethylamine, 7.8eq. of POCl 3 heating at 90 0 C for 2 h.
  • This step involves the displacement of a leaving group (LG), preferably chlorine, in formula (II) with an amino group of formula (IV) in a suitable solvent (such as isopropanol or dioxane) in the presence of an organic base (such as N,N-diisopropylethylamine) at temperatures in the range of 0- 80 0 C for reaction times of up to 24 h.
  • LG leaving group
  • an organic base such as N,N-diisopropylethylamine
  • Scheme 2 illustrates the conversion of compounds with formula (V) to compounds with formula (VI), wherein Rl, R2, R3, R7 and LG are as defined previously.
  • Compounds of formula (V) can be converted to compounds with formula (VI) by displacement of a leaving group (preferably chlorine) in formula (V) with an amine with formula (VII) in an appropriate solvent (e.g. acetonitrile or n-butanol) in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid).
  • a suitable organic acid or mineral acid such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid.
  • the reaction can be carried out at temperatures ranging from 50 0 C to 200 0 C by convectional heating or microwave heating.
  • Preferred method leq. of formula (V), leq. of acetic acid, 3eq. of formula (
  • compounds of formula (VI) can be prepared from compounds with formula (V) (where LG is chlorine) by reaction with amine (VII) in the presence of a palladium source (e.g. Pd(OAc) 2 or Pd 2 (dba) 3 ), a suitable ligand (e.g. bis(diphenylphosphino)-9,9-dimethylxanthene) and a suitable base (e.g. Cs 2 CO 3 or sodium tert-butoxide) in a suitable solvent (e.g. dioxane).
  • a palladium source e.g. Pd(OAc) 2 or Pd 2 (dba) 3
  • a suitable ligand e.g. bis(diphenylphosphino)-9,9-dimethylxanthene
  • a suitable base e.g. Cs 2 CO 3 or sodium tert-butoxide
  • solvent e.g. dioxane
  • Preferred method leq. of formula (V), 1.2eq. of amine (VII), 0.06eq. of Pd 2 (dba) 3 , 0.12eq. of bis(diphenylphosphino)-9,9-dimethylxanthene, 3eq. of sodium tert-butoxide in dioxane at 105 0 C for 18 h.
  • Compounds of formula (IX) and formula (X) can be prepared from compounds of formula (VIII) according to scheme 3, wherein Rl, R2, R7 and LG are as defined previously; Q is NR7 or oxygen and PG represents a suitable protecting group (such as tert-butoxycarbonyl or benzyloxycarbonyl).
  • Compounds of formula (IX) and formula (X) can be prepared from compounds of formula (VIII) by displacement of a leaving group (preferably chlorine) in formula (VIII) with an amine with formula (VII) in an appropriate solvent (e.g.
  • acetonitrile or n-butanol in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid).
  • a suitable organic acid or mineral acid such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid.
  • the reaction can be carried out at temperatures typically ranging from 50 0 C to 200 0 C, either by convectional heating or microwave heating. It will be understood by a skilled person that certain protecting groups are sensitive to the reaction conditions and may therefore give the product in the unprotected form (formula X), or indeed give mixtures of both formula (IX) and formula (X).
  • Preferred conditions leq of formula (VIII), 1.3 eq of formula (VII), 1.45eq of 4M HCl in dioxane, in aqueous acetonitrile at 50 0 C overnight.
  • Compounds of formula (X) can be prepared from compounds of formula (IX) using standard methods for amine deprotection known to a skilled person. For example, where PG is tert- butoxycarbonyl reaction of formula (IX) with a suitable acid (such as HCl or trifluoroacetic acid) in a suitable solvent (such as dioxane or DCM) at temperatures typically in the range of 0 0 C to the reflux temperature of the solvent enables this transformation to occur. Preferred conditions: Formula (IX) is stirred in 4M HCl in dioxane at room temperature for 2 h.
  • a suitable acid such as HCl or trifluoroacetic acid
  • a suitable solvent such as dioxane or DCM
  • the reaction is carried out in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine) in a suitable solvent (such as DCM or dioxane) at temperatures ranging from O 0 C to the reflux temperature of the solvent Preferred conditions: leq of formula (X), 1.02eq of formula (XII), 2.2eq of triethylamine in DCM at room temperature for 2 h.
  • a suitable base such as N,N-diisopropylethylamine or triethylamine
  • a suitable solvent such as DCM or dioxane
  • compounds with formula (XI) can be prepared from compounds of formula (X) by treatment with formula (XII), where X is hydroxy.
  • the reaction is carried out in the presence of a suitable amine coupling reagent known to the skilled person, (such as but not limited to HATU, DCC, EDCI), optionally in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine), optionally in the presence of an additive (such as but not limited to, 1- hydroxybenzotriazole) in a suitable solvent (such as DMF or pyridine) at temperatures ranging from 0 0 C to the reflux temperature of the solvent.
  • a suitable amine coupling reagent known to the skilled person, such as but not limited to HATU, DCC, EDCI
  • a suitable base such as N,N-diisopropylethylamine or triethylamine
  • an additive such as but not limited to, 1- hydroxybenzotriazo
  • Preferred conditions leq of formula (X), l. ⁇ eq of formula (XII), 1.7eq of HATU, 6.2eq of N,N- diisopropylethylamine in DMF at room temperature overnight.
  • compounds with formula (XI) can be prepared according to Scheme 5 wherein Rl, R2, R6 and R7, LG are all as defined previously; X is chlorine or hydroxyl.
  • Compounds with formula (VIII), wherein PG is tert-butoxycarbonyl and LG is preferably chlorine are converted to formula (XIII) on treatment with a suitable acid (such as HCl or trifluoroacetic acid) in a suitable solvent (such as dioxane or DCM) at temperatures typically in the range of 0 0 C to the reflux temperature of the solvent. Typical reaction times are in the range of 1 h to 24 h.
  • a suitable acid such as HCl or trifluoroacetic acid
  • a suitable solvent such as dioxane or DCM
  • Typical reaction times are in the range of 1 h to 24 h.
  • Formula (VIII) is stirred in 4M HCl in dioxane at room temperature for 1 h.
  • Compounds with formula (XIV) can be prepared from compounds of formula (XIII) by treatment with formula (XII), wherein X is chlorine.
  • the reaction is carried out in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine) in a suitable solvent (such as DCM or dioxane) at temperatures ranging from 0 0 C to the reflux temperature of the solvent.
  • a suitable base such as N,N-diisopropylethylamine or triethylamine
  • a suitable solvent such as DCM or dioxane
  • compounds with formula (XIV) can be prepared from compounds of formula (XIII) by treatment with formula (XII), wherein X is hydroxyl.
  • the reaction is carried out in the presence of a suitable amine coupling reagent known to the skilled person, (such as but not limited to HATU, DCC, EDCI), optionally in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine), optionally in the presence of an additive (such as but not limited to, hydroxybenzotriazole) in a suitable solvent (such as DMF or pyridine) at temperatures ranging from O 0 C to the reflux temperature of the solvent.
  • a suitable amine coupling reagent known to the skilled person, such as but not limited to HATU, DCC, EDCI
  • a suitable base such as N,N-diisopropylethylamine or triethylamine
  • an additive such as but not limited to, hydroxybenzotriazo
  • Compounds of formula (XIV) can be converted to compounds with formula (XI) by displacement of a leaving group (preferably chlorine) in formula (XIV) with an amine with formula (VII) in an appropriate solvent (e.g. acetonitrile or n-butanol) in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid).
  • a suitable organic acid or mineral acid such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid.
  • the reaction can be carried out at temperatures typically ranging from 50 0 C to 200 0 C, by either convectional heating or optionally with microwave heating.
  • Pd(OAc) 2 or Pd 2 (dba) 3 a suitable ligand (e.g. bis(diphenylphosphino)-9,9-dimethylxanthene) and a suitable base (e.g. Cs 2 CO 3 or sodium terf-butoxide) in a suitable solvent (e.g. dioxane).
  • a suitable solvent e.g. dioxane
  • Preferred method leq. of formula (XIV), 1.2eq. of amine (VII), 0.06eq. of Pd 2 (dba) 3 , 0.12eq. of bis(diphenylphosphino)-9,9-dimethylxanthene, 3eq. of sodium tert-butoxide in dioxane at 105 0 C for 18 h.
  • Compounds with formula (XVI) can be prepared from compounds of formula (XV) according to Scheme 6, wherein Rl, R2, R4, R5 and R6 are as defined previously.
  • Compounds of formula (XV) can be prepared according to step (c), scheme 2.
  • Compounds of with formula (XVI) can be prepared from compounds of formula (XV) by treatment with a suitable amine with the formula NHR4R5.
  • the reaction is carried out in the presence of a suitable amine coupling reagent known to the skilled person, (such as but not limited to HATU, DCC, EDCI), optionally in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine), optionally in the presence of an additive (such as but not limited to, 1- hydroxybenzotriazole) in a suitable solvent (such as DMF or pyridine) at temperatures ranging from 0 0 C to the reflux temperature of the solvent.
  • a suitable amine coupling reagent known to the skilled person, (such as but not limited to HATU, DCC, EDCI)
  • a suitable base such as N,N-diisopropylethylamine or triethylamine
  • an additive such as but not limited to, 1-
  • a further aspect of the invention relates to a process for preparing a compound of formula XIX, wherein R 1 , R 2 and R 3 are as defined above, said process comprising the steps of:
  • Compounds with formula XVIII can be prepared by reacting compounds with formula (XVII) with compounds with formula (II) in a suitable solvent (such as THF) in the presence of a strong base, such as sodium hydride at temperatures ranging from 0 0 C to the reflux temperature of the solvent.
  • a strong base such as sodium hydride
  • leq. of formula (XVII) is deprotonated with leq. of sodium hydride in THF at O 0 C, followed by treatment with leq. of formula (II) at room temperature overnight.
  • Compounds of formula (XVIII) can be converted to compounds with formula (XIX) by displacement of a leaving group (preferably chlorine) in formula (XVIII) with an amine with formula (VII) in an appropriate solvent (e.g. n-butanol) in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid).
  • a suitable organic acid or mineral acid such as acetic acid, HCl, H 2 SO 4 , p-toluenesulfonic acid.
  • the reaction can be carried out at temperatures typically ranging from 50 0 C to 200 0 C, by either convectional heating or optionally with microwave heating.
  • Preferred method leq. of formula (XVIII), 0.2eq. of acetic acid, 2eq. of formula (IV) in n-butanol heated to 150 0 C in the microwave for 15 minutes.
  • GST glutathione S-transferase
  • His ⁇ hexahistidine
  • E.coli The following proteins were expressed in E.coli:- CHK2[5-543], cyclin-dependent protein kinase 2 (CDK2), MAP kinase-interacting kinase 2 (MNK2), extra-cellular signal-regulated kinase 1 (ERKl).
  • CDK2 cyclin-dependent protein kinase 2
  • MNK2 MAP kinase-interacting kinase 2
  • ERKl extra-cellular signal-regulated kinase 1
  • kinases were expressed in Sf21 cells:Aurora B and Aurora C, extra-cellular signal- regulated kinase 8 (ERK8), microtubule affinity regulating kinase 3 (MARK3), protein kinase B ⁇ [118-480][S473D], protein kinase B ⁇ (PKB ⁇ [120-481][S474D], 3-phosphoinositide-dependent protein kinase- 1 [52-556]
  • insect Sf21 cells were incubated for I h with the protein phosphatase inhibitor okadaic acid (50 nM). JNK isoforms were activated with
  • MKK4 and MKK7 MNK2 with p38 ⁇ MAPkinase; PKB ⁇ , PKB ⁇ , with PDKl, and ERKl with
  • PreScission protease and the CDK2-cyclin A2 complex purified by chromatography on SP- Sepharose. It was then activated with CAK1/CDK7 followed by chromatography on nickel- nitrilotriacetate agarose to remove CAK1/CDK7, which binds to this column by virtue of its C- terminal His6 tag. All the other protein kinases were active as expressed.
  • Protein kinase assays All assays (25.5 ⁇ l) were carried out at room temperature (21°C) and were linear with respect to time and enzyme concentration under the conditions used. Assays were performed for 30 min using Multidrop Micro reagent dispensers (Thermo Electron Corporation, Waltham, MA 02454, USA) in a 96- well format. The concentration of magnesium acetate in the assays was 10 mM and the [ ⁇ - 33P] ATP (800 cpm / pmol) was used at 5, 20 or 50 ⁇ M as indicated, in order to be at or below the Km for ATP for each enzyme.
  • Multidrop Micro reagent dispensers The concentration of magnesium acetate in the assays was 10 mM and the [ ⁇ - 33P] ATP (800 cpm / pmol) was used at 5, 20 or 50 ⁇ M as indicated, in order to be at or below the Km for ATP for each enzyme.
  • Protein kinases assayed at 5 ⁇ M ATP were:- ERKl, ERK8, PKB ⁇ , MARK3, Aurora C. Protein kinases assayed at 20 ⁇ M ATP were:- JNKl, PDKl 5 CHKl, CHK2, CDK2 and Aurora B. Protein kinases assayed at 50 ⁇ M ATP were:-MNK2, IKKepsilon and TBKl .
  • the assays were initiated with MgATP, stopped by addition of 5 ⁇ l of 0.5 M orthophosphoric acid and spotted on to P81 filterplates using a unifilter harvester (PerkinElmer, Boston, MA 02118, USA). The IC50 values of inhibitors were determined after carrying out assays at 10 different concentrations of each compound.
  • ERKl and ERK8 were both assayed against myelin basic protein (MBP, 0.33mg/ml).
  • MARK3 was assayed against the peptide KKKVSRSGL YRSPSMPENLNRPR (300 ⁇ M), MNK2 against the eIF4E protein (0.5mg/ml).
  • PKB ⁇ was assayed against the peptide GRPRTSSFAEGKK (30 ⁇ M).
  • TBKl were assayed against (AKPKGNKD YHLQTCCGSLA YRRR) (300 ⁇ M).
  • the substrates used for other protein kinases were described previously. 8 ' 9
  • enzymes were diluted in 50 mM Tris/HCl pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA, 0.1% (v/v) 2-mercaptoethanol and assayed in 50 mM Tris/HCl pH 7.5, 0.1 mM EGTA, 0.1 % (v/v) 2-mercaptoethanol.
  • Preparative high pressure liquid chromatography was carried out using apparatus made by Agilent.
  • the apparatus is constructed such that the chromatography (column: either a 30 x 100 mm (10 ⁇ m) C-18 Phenomenex Gemini column at a flow rate of 50 ml/min, or a 21.2 x 100 mm (5 ⁇ m) C-18 Phenomenex Gemini column at a flow rate of 20 ml/min) is monitored by a multi-wavelength UV detector (G1365B manufactured by Agilent) and an MM-ES+APCI mass spectrometer (G-1956A, manufactured by Agilent) connected in series, and if the appropriate criteria are met the sample is collected by an automated fraction collector (G1364B manufactured by Agilent).
  • Flash chromatography refers to silica gel chromatography and carried out using an SP4 MPLC system (manufactured by Biotage); pre-packed silica gel cartridges (supplied by Biotage); or using conventional glass column chromatography.
  • TLC thin layer chromatography
  • R f is the distance travelled by the compound divided by the distance travelled by the solvent on a TLC plate.
  • Compound preparation Where the preparation of starting materials is not described, these are commercially available, known in the literature, or readily obtainable by those skilled in the art using standard procedures. Where it is stated that compounds were prepared analogously to earlier or later examples or intermediates, it will be appreciated by the skilled person that the reaction time, number of equivalents of reagents and temperature can be modified for each specific reaction and that it may be necessary or desirable to employ different work-up or purification techniques.
  • the microwave used is an Initiator 60 supplied by Biotage. The actual power supplied varies during the course of the reaction in order to maintain a constant temperature.
  • Xantphos 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene
  • HATU N,N,N l ,N'-Tetramethyl-0-(7-azabenzotriazol- 1 -yl)uronium- hexafluorophospate
  • EDCI 1,3-Propanediamine, N3-(ethylcarbonimidoyl)-Nl, Nl -dimethyl-, hydrochloride
  • DIPEA N,N-diisopropylethylamine
  • Alternatively intermediate 8 can be prepared according to the following method:
  • N-(3-nitrophenyl)pyrrolidine-l-carboxamide prepared in step 1 (10 g, 42.6 mmol) in ethyl acetate (150 ml) and ethanol (10 ml) was added 10% Pd on activated carbon (1 g). The reaction was stirred under an atmosphere of hydrogen at room temperature for 18 h. More 10% Pd on activated carbon (0.4 g) was added and the reaction was complete after stirring for a further 4 hours. The mixture was filtered through CeliteTM, washing with ethanol, and then evaporated to give a pink foam, which was triturated with petroleum ether - EtOAc (10:1).
  • N,N,N'-Trimethylethylenediamine (798 ⁇ l, 6.29 mmol) was added to 2-chloro-5-nitropyridine (1.0 g, 6.29 mmol) and DIPEA (1.09 ml, 6.29 mmol) in MeCN (20 ml). The mixture was stirred at rt for
  • Step l ⁇ -fCyclobutanecarbonyl-amino) -propyl] '-methyl-carbamic acid tert-butyl ester
  • N-(3-Aminopropyl)-N-methylcarbamic acid tert-butylester 600 mg, 3.19 mmol
  • N,N-diisopropylethylamine 610 ⁇ l, 3.51 mmol
  • cyclobutanecarbonyl chloride 366 ⁇ l, 3.19 mmol
  • the solution was cooled to 0 0 C and then quenched with saturated sodium hydrogen carbonate(aq) and the aqueous phase extracted twice with DCM.
  • the combined organic extracts were washed with brine and the solution filtered through a phase separation cartridge and solvents evaporated.
  • the crude product was purified by flash chromatography using a Biotage SP4 (ethyl acetate/methanol gradient) to give the product (165 mg, 14%).
  • 2-Oxo-pyrrolidine-l-carboxylic acid tert-butyl ester (2 g, 1-0.8 mmol) was dissolved in anhydrous THF (100 ml), under nitrogen, cooled down to -78 0 C and to it added LHMDS (1 M solution in THF, 32.4 ml, 32.4 mmol) dropwise. The solution was left to stir at -78 0 C for 45 min. prior to dropwise addition of methyl iodide (4.0 ml, 64.9 mmol). The resulting mixture was stirred at -78 0 C for a further 30 min. and at then room temperature overnight. The solution was then cooled down to 0 0 C then quenched with water, followed by IM HCl.
  • the aqueous phase was extracted with diethyl ether (x3) and the combined organic extracts washed with saturated sodium hydrogen carbonate, followed by brine, dried (MgSO 4 ) and the solvent evaporated.
  • the crude material was purified by flash chromatography using a Biotage SP4 (petroleum ether/ethyl acetate gradient) to give the product as a brown solid (1.0 g, 43%).
  • Example 70 120 mg, 0.28 mmol was added 30%
  • Example 70 Thionyl chloride (253 ⁇ l, 3.5 mmol) was added dropwise to a suspension of Example 70 (300 mg, 0.69 mmol) in DCM (25 ml) at 0 0 C. The mixture was then allowed to warm to room temperature and stirred for 18 h producing a white precipitate. The reaction was evaporated to give the desired product as a white solid (100%).
  • Iron powder (517 mg, 9.27 mmol) was added to a solution of 3-nitrophenylacetonitrile (500 mg, 3.09 mmol) in acetic acid (9.5 ml) and water (6.6 ml). The reaction was stirred at 40 0 C for 24 h, then cooled to room temperature. The mixture was diluted with water (20 ml), adjusted to pH 8 with 2N NaOH and then filtered trough Celite®. The aqueous filtrate was extracted with ethyl acetate, the combined organic phases were dried (MgSO,)) and evaporated.
  • N-(2-chloroethyl)morpholine HCl salt (2.1 g, 11.06 mmol) was added portionwise to a stirred mixture of 4-nitro-lH-pyrazole (1.0 g, 8.85 mmol) and KOH (1.24 g, 22.12 mmol) in EtOH (20 ml). The mixture was heated under reflux for 2 h and allowed to cool to rt. After dilution with EtOAc and water the organic phase was washed with brine, dried and concentrated. The residue was purified by flash column chromatography on silica gel (100 g) eluting with 50:1 DCM-MeOH to provide an orange oil (987 mg, 49 %).
  • 6-Bromo-[l,2,4]triazolo[l,5-a]pyridine 300 mg, 1.5 mmol
  • bezophenone imine 326 mg, 1.8 mmol
  • Pd 2 (dba) 3 7 mg, 0.008 mmol
  • BINAP 14 mg, 0.02 mmol
  • sodium terf-butoxide 202 mg, 2.1 mmol
  • Biotage SP4 ethyl acetate / petroleum ether gradient
  • the first eluting product was l-cyclopropylmethyl-6-nitro-lH-indazole (Intermediate 71) obtained as a yellow solid (1.14 g, 43%).
  • 1 H NMR 400 MHz, CHLOROFORM- d
  • ppm 0.42 - 0.54 m, 2 H
  • 0.59 - 0.72 m, 2 H
  • 1.33 - 1.44 m, 1 H
  • 7.86 (d, / 8.24 Hz, 1 H)
  • 7.99 - 8.06 m, 1 H
  • Step 2 1 -Cyclopropylmethyl- lH-pyrazol-4-ylamine
  • Examples 2-10 were prepared analogously to Example 1 (the general structure is shown below followed by the tabulated examples).
  • Examples 12-25 were prepared analogously to example 11 (the general structure is shown below followed by the tabulated examples).
  • Examples 33-36 were prepared analogously to example 32 (the general structure is shown below followed by the tabulated examples).
  • Example 34 To a solution of Example 34 (20.1 mg, 0.036 mmol) in DMF (0.5 ml) was added 1- methylpiperazine (6 ⁇ l, 0.055 mmol), HATU (22 mg, 0.58 mmol) and ⁇ , ⁇ -diisopropylethylamine (38 ⁇ l, 0.219 mmol). The reaction was stirred at room temperature for 18 hours. The mixture was evaporated then filtered through a 1 g Isolute-NH 2 cartridge, eluting with 9:1 DCM : methanol. Purification by flash chromatography on the Biotage SP4 (gradient elution from 0 to 10% methanol in DCM) gave the desired product as a white solid (18.9 mg, 98%).
  • Examples 54-55 were prepared analogously to Example 53 (the general structure is shown below followed by the tabulated examples).

Abstract

A first aspect of the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt or ester thereof, formula (I): wherein: R1 is C3-8-cycloalkyl; X is O, NR7 or C3-6-heterocycloalkyl; R2 is aryl, heteroaryl, fused or unfused aryl-C3-6-heterocycloalkyl or fused or unfused heteroaryl-C3-6-heterocycIoalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, C1-6-alkyl, C3-7-cycloalkyl and a group A, wherein said C1-6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl, R10 and a group A, said heteroaryl group is optionally substituted by one or more R10 groups; and wherein said C3-6-heterocycloalkyl group optionally contains one or more groups selected from oxygen, sulfur, nitrogen and CO; R3 is C1-6-alkyl optionally substituted by one or more substituents selected from aryl, heteroaryl, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7SO2R6, -NR7COOR7, -CONR4R5, C3-6-heterocycloalkyl and formula (a, b, c): wherein R4-7 and A are as defined in the claims. Further aspects relate to the use of said compounds in the treatment of various therapeutic disorders, and more particularly as inhibitors of one or more kinases.

Description

PYRIMIDINE DERIVATIVES CAPABLE OF INHIBITING ONE OR MORE KINASES
The present invention relates to pyrimidine compounds that are capable of inhibiting one or more kinases. The compounds find applications in the treatment of a variety of disorders, including cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, primary open angle glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and/or chronic inflammation.
BACKGROUND TO THE INVENTION Pyrimidines and analogues thereof are already described as active ingredients, such as, for example, the 2-anilino-pyrimidines as fungicides (DE-A-4029650) or substituted pyrimidine derivatives for treating neurological or neurodegenerative diseases (WO 99/19305). As CDK inhibitors, the most varied pyrimidine derivatives are described, for example, bis(anilino)- pyrimidine derivatives (WO 00/12486), 2-amino-4-substituted pyrimidines (WO 01/14375), purines (WO 99/02162), 5-cyano-pyrimidines (WO 02/04429), anilinopyrimidines (WO 00/12486) and 2-N-dimethylaminopropoxy-pyrimidines (WO 00/39101).
It is amongst the objects of the present invention to provide compounds which display a high degree of activity and/or specificity to particular kinases and may therefore serve as drug candidates or as starting points for further derivatisation and kinase inhibition studies.
It is a further object of the present invention to provide compounds for potential use as drug candidates for treating cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, inflammatory disease and/or primary open angle glaucoma (POAG).
It is a further object to provide compounds which display a significant inhibitory effect on one or more of the following kinases: TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and/or IKKepsilon.
STATEMENT OF INVENTION
A first aspect of the invention relates to a compound of formula (I), or a pharmaceutically acceptable salt or ester thereof,
Figure imgf000003_0001
(I) wherein: R1 is C3-8-cycloalkyl;
X is O, NR7or C^-heterocycloalkyl;
R2 is aryl, heteroaryl, fused or unfused aryl-C^-heterocycloalkyl or fused or unfused heteroaryl-C3. 6-heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci-6-alkyl, C3-7-cycloalkyl and a group A, wherein said
Figure imgf000003_0002
group is optionally substituted by one or more substituents selected from aryl, heteroaryl, R10 and a group A, said heteroaryl group is optionally substituted by one or more R10 groups; and wherein said C3-6- heterocycloalkyl group optionally contains one or more groups selected from oxygen, sulfur, nitrogen and CO;
R3 is Ci_6-alkyl optionally substituted by one or more substituents selected from aryl, heteroaryl, -NR > 44τR> 53,
Figure imgf000003_0003
- vNττR> 7XCO)NR4R', -NR7SO2R6, -NR 'COOR', -CONR >44rR> 55, C3-6-heterocycloalky] and
Figure imgf000003_0004
wherein said aryl, heteroaryl and C^-heterocycloalkyl groups are each optionally substituted by one or more substituents selected from -Ci.6-alkyl and a group A, wherein said -C^-alky! group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A; A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -NO2, -NH2, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7COOR7, -NR7(SO2)R6, - CO2H, -NR7(SO2)NR4R5, -COOR7, -CONR4R5, COR6, SO2NR4R5 and -SO2R6;
each R4 and R5 is independently selected from hydrogen, C3-7-cycloalkyl, aryl, heteroaryl, Ci-β-alkyl and a C3^-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more R10 groups, wherein said C^-alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR8R9, -NR7(CO)R6, NR7COOR6, -NR7(SO2)R6, -COOR6, -CONR8R9, OR10, -SO2R6 and a C^-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more or R10 groups; or
R4 and R5 together with the N to which they are attached form a
Figure imgf000004_0001
ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C^-heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR8R9 and R1D groups;
each R6 is independently selected from C^-alkyl, C3-7 cycloalkyl, C4.7-heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9;
each R7 is selected from hydrogen,
Figure imgf000004_0002
is optionally substituted by one or more halogens;
each of R8 and R9 is independently selected from hydrogen and Ci_5-alkyl, wherein said Ci^-alkyl group is optionally substituted by one or more halogens; or
R8 and R9 together with the N to which they are attached form a C4-6-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C^-heterocycloalkyl ring is optionally substituted by one or more R10 groups; and each R10 is selected from halogen, C3.7-cycloalkyl and C]-6-alkyl optionally substituted by one or more halogens, wherein where R10 is C^-alkyl and two or more R10 groups are attached to the same carbon atom, the R10 groups may be linked to form a spiroalkyl group.
As is demonstrated in Examples section that follows, representative compounds of the present invention were tested for their kinase inhibition activity and showed significant potency to TBKl and/or MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and/or IKKepsilon. These compounds can therefore efficiently serve for treating diseases or disorders in which inhibiting the activity of one or more of these kinases, would be beneficial. When the kinase is PDKl, the present invention relates to the treatment of diseases where it would be desirable to inhibit PDKl and at least one other identified kinase.
A second aspect of the invention relates to a pharmaceutical composition comprising at least one compound as described above and a pharmaceutically acceptable carrier, diluent or excipient.
A third aspect of the invention relates to a compound as described above for use in medicine.
A fourth aspect of the invention relates to a compound as described above for use in treating a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
A fifth aspect of the invention relates to the use of a compound as described above in the preparation of a medicament for treating or preventing a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, primary open angle glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
A sixth aspect of the invention relates to the use of a compound as described above in the preparation of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity, wherein the kinase is selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2,
MARK3, MELK, IRR, VEG-FR, IKKepsilon and combinations thereof.
A seventh aspect of the invention relates to a method of treating a mammal having a disease state alleviated by the inhibition of a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound as described above.
An eighth aspect of the invention relates to the use of a compound as described above in an assay for identifying further candidate compounds capable of inhibiting one or more kinases selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon.
A ninth aspect of the invention relates to a process for preparing a compound of formula IV, wherein R1, R2, R3 and R7 are as defined above, said process comprising the steps of:
Figure imgf000006_0001
(X=O1 S)
Ia II III IV
(i) converting a compound of formula Ia to a compound of formula II, where each LG is independently a leaving group; (ii) reacting said compound of formula II with an amine of formula IV to form a compound of formula III;
(iii) reacting said compound of formula III with an amine of formula VII to form a compound of formula IV. A tenth aspect of the invention relates to a process for preparing a compound of formula XIX, wherein R1, R2 and R3 are as defined above, said process comprising the steps of:
Figure imgf000007_0001
(X=O, S) Ia II XVIII XIX
(i) converting a compound of formula Ia to a compound of formula II, where each LG is independently a leaving group; (ii) reacting said compound of formula II with an amine of formula XVII to form a compound of formula XVIII; (iii) reacting said compound of formula XVIII with an amine of formula VII to form a compound of formula XIX.
DETAILED DESCRIPTION
"Alkyl" is defined herein as a straight-chain or branched alkyl radical, for example, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, hexyl. Preferably, the alkyl group is a C1-6 alkyl group, more preferably a CM group.
"Cycloalkyl" is defined herein as a monocyclic alkyl ring, such as, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. Preferably, the cycloalkyl group is a C3.12 cycloalkyl group, more preferably a C3-6 cycloalkyl group.
"Halogen" is defined herein as chloro, fluoro, bromo or iodo.
As used herein, the term "aryl" refers to a C6-12 aromatic group, which may be benzocondensed, for example, phenyl or naphthyl. "Heteroaryl" is defined herein as a monocyclic or bicyclic C2-12 aromatic ring comprising one or more heteroatoms (that may be the same or different), such as oxygen, nitrogen or sulfur. Examples of suitable heteroaryl groups include thienyl, furanyl, pyrrolyl, pyridinyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl etc. and benzo derivatives thereof, such as benzofuranyl, benzothienyl, benzimidazolyl, indolyl, isoindolyl, indazolyl etc.; or pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl etc. and benzo derivatives thereof, such as quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl etc.
"Heterocycloalkyl" refers to a cyclic aliphatic group containing one or more heteroatoms selected from nitrogen, oxygen and sulfur, which is optionally interrupted by one or more -(CO)- groups in the ring and/or which optionally contains one or more double bonds in the ring. Preferably, the heterocycloalkyl group comprises 3-6 carbon atoms and is fully saturated. Preferred heterocycloalkyl groups include piperidinyl, pyrrolidinyl, piperazinyl, thiomorpholinyl and morpholinyl. More preferably, the heterocycloalkyl group is selected from N-piperidinyl, N- pyrrolidinyl, N-piperazinyl, N-thiomorpholinyl and N-morpholinyl.
In one preferred embodiment of the invention, R1 is cyclopropyl or cyclobutyl.
More preferably, R1 is cyclopropyl.
In one preferred embodiment of the invention, X is O or NR7, more preferably, O, NMe or NH. Even more preferably, X is NH.
In one preferred embodiment, A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -NO2, -NH2, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7COOR7, -NR7(SO2)R6, - CO2H, -NR7(SO2)NR4R5, -COOR7, -CONR4R5, COR6 and -SO2CH3.
In one preferred embodiment of the invention, R2 is an optionally substituted aryl or heteroaryl group selected from the following:
Figure imgf000009_0001
Figure imgf000009_0002
Preferably, the substituent attached to the aryl or heteroaryl group is selected from
Figure imgf000009_0003
C3-7- cycloalkyl and a group A, wherein said
Figure imgf000009_0004
group is in turn optionally substituted by one or more substituents selected from aryl, heteroaryl, R10 and a group A.
In one particularly preferred embodiment of the invention, R2 is an optionally substituted aryl or heteroaryl group selected from the following:
Figure imgf000010_0001
In a more preferred embodiment, R2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from CJ-6-alkyl and a group A, wherein said Ci-6- alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A, and wherein A is selected from halogen, OH, CN, CF3, -NH2, -NR4R5, -OR6, NR7(CO)R6, -NR7COOR7, -NR7(SO2)R6, -COOH, -COOR7 and CONR4R5.
In an even more preferred embodiment, R2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from
Figure imgf000010_0002
halogen, CN, NHCO-
C^-alkyl, CF3, COOH, CONH2, OH, NH2, NHSO2-C M-alkyl, 0-CF3, -NHCOO-C ,^-alkyl, -CO2-
Ci_5-alkyl, -N(CI-6-alkyl)2, 4-methylpiperazin-l-yl, (4-methylpiperazin-l-yl)-C0-, (N- morpholiny])-(CH2)p(0)q-, (imidazol-l-yl)-(CH2)p- where q is O, 1, 2 or 3 and each p is independently 1 , 2 or 3 and NR4R5, wherein R4 and R5 and the nitrogen to which they are attached form a C^-heterocycloalkyl ring optionally containing a CO group.
More preferably, R2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from Me, Cl, F, CN, NHCOMe, CF3, COOH, CONH2, OH, NH2, NHSO2Me, OCF3, -NHCOO1Bu, -CO2Me, -NMe2, 4-methylpiperazin-l-yl, N-morpholinyl, (4- methylpiperazin- 1 -yl)-CO-, (N-morpholinyl)-CH2CH2O-, (imidazol- 1 -yl)-CH2- and
Figure imgf000010_0003
In one preferred embodiment, R2 is a phenyl group optionally substituted by one or more groups selected from C]-6-alkyl, heteroaryl and A, wherein said
Figure imgf000011_0001
and heteroaryl groups are in turn optionally substituted.
In one preferred embodiment, R2 is a phenyl group substituted by one or more A groups. Preferably, the A group is selected from CF3, halogen, CN, NHSO2Me, SO2NR4R5, NR4R5, OR6, COOR7, NR7COOR7, NR7COR6, CONR4R5, NR7CONR4R5 and NR7SO2R6.
In one preferred embodiment, R2 is a phenyl group substituted by one or more
Figure imgf000011_0002
groups, each of which in turn is optionally substituted by one or more groups selected from heteroaryl and A. Preferably, the heteroaryl group is selected from imidazolyl and triazolyl. Preferably, the A group is selected from CONR4R5, NR4R5, OR6, COOR7 and CN.
In one preferred embodiment, R2 is a phenyl group substituted by one or more heteroaryl groups. Preferably, the heteroaryl group is selected from pyrimidinyl, tetrazolyl, pyridinyl, pyrazolyl, oxazolyl and triazolyl.
In one preferred embodiment, R2 is a pyridyl group optionally substituted by one or more groups selected from
Figure imgf000011_0003
heteroaryl and A, wherein said
Figure imgf000011_0004
and heteroaryl groups are in turn optionally substituted.
In one preferred embodiment, R2 is a pyridyl group substituted by one or more A groups. Preferably, the A group is selected from NR4R5, halo and OR6.
In one preferred embodiment, R2 is a pyridyl group substituted by a heteroaryl group. Preferably, the heteroaryl group is selected from pyrazolyl, pyrimidinyl and pyridinyl.
In one preferred embodiment, R2 is a pyridyl group substituted by a Ci-β-alkyl group, wherein said Ci-6-alkyl group is in turn optionally substituted with one or more substituents selected from NR4R5 and OR6. In one preferred embodiment, R2 is an optionally substituted fused aryl-Cs^-heterocycloalkyl or fused heteroaryl-C3^-heterocycloalkyl. Preferably, R2 is selected from the following:
Figure imgf000012_0001
Preferably, the substituent is an optionally substituted
Figure imgf000012_0002
group.
In one preferred embodiment of the invention, R3 is C^-alkyl optionally substituted by one or more substituents selected from heteroaryl, -NR4R5, -NR7(CO)R6, -NR7COOR7, C3^- heterocycloalkyl and
Figure imgf000012_0003
In one highly preferred embodiment, R3 is Ci-4-alkyl substituted by -NR7(CO)R6. Preferably, for this embodiment, R7 is H and R6 is selected from C^-alkyl, C3.7 cycloalkyl, C4-7-heterocycloalkyl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9.
In one preferred embodiment, R3 is CM-alkyl substituted by -NR7(CO)R6 and R6 is selected from thienyl, cyclopentyl, CH2-cyclopentyl, methyl, isopropyl, n-propyl, pyrazolyl, cyclohexyl, thiazolyl, oxazolyl, furanyl, imidazolyl, cyclopropyl, CH2-cyclopropyl, cyclobutyl, triazolyl, pyrrolyl, tetrahydrofuranyl and isoxazolyl, each of which may be substituted by one or more R10 or -NR8R9 groups. In one preferred embodiment, R3 is CM-alkyl substituted by -NR7(CO)R6 and R6 is selected from thienyl, cyclopentyl, CH2-cyclopentyl, methyl, isopropyl, n-propyl, pyrazolyl, cyclohexyl, thiazolyl, oxazolyl, furanyl, imidazolyl, cyclopropyl, CH2-cyclopropyl, cyclobutyl, triazolyl, pyrrolyl, tetrahydrofuranyl and isoxazolyl, each of which may be substituted by one or more halogen or Ci-6- alkyl groups.
Even more preferably, R3 is
Figure imgf000013_0001
substituted by -NR7(CO)R6 and R6 is selected from thienyl, cyclopentyl, CH2-cyclopentyl, methyl, isopropyl, n-propyl, pyrazolyl, cyclohexyl, thiazolyl, oxazolyl, furanyl, CF3, imidazolyl, cyclopropyl, CH2-cyclopropyl, cyclobutyl, triazolyl, pyrrolyl, tetrahydrofuranyl, CH2NMe2 and isoxazolyl.
In another highly preferred embodiment, R3 is
Figure imgf000013_0002
substituted by a heteroaryl group. Preferably, the heteroaryl group is selected from pyrazolyl, tetrazolyl and triazolyl.
In another highly preferred embodiment, R3 is CM-alkyl substituted by a -NR4R5 group. Preferably, R4 and R5 together with the N to which they are attached form a C^-heterocycloalkyl ring optionally further containing one or more CO groups, wherein said C^-heterocycloalkyl ring is optionally substituted with one or more R10 groups. Preferably for this embodiment, R10 is Ci-5- alkyl optionally substituted with one or more halo atoms.
In one preferred embodiment, R3 is CM-alkyl substituted by one of the following groups:
Figure imgf000013_0003
each of which may be optionally substituted with one or more R groups In one highly preferred embodiment, R3 is Ci^-alkyl substituted by one of the following groups:
Figure imgf000014_0001
In another highly preferred embodiment, R3 is CM-alkyl substituted by a Cs-β-heterocycloalkyl group, wherein said Cs-β-heterocycloalkyl group is optionally substituted by one or more A groups. Preferably, for this embodiment, the A group is a COR6 group.
In another highly preferred embodiment, R3 is C^-alkyl substituted by a group selected from:
Figure imgf000014_0002
In one especially preferred embodiment of the invention, R is an optionally substituted C3-alkyl group.
In one particularly preferred embodiment, the compound of the invention is selected from the following: 9 10 11 12 13 14 15 16
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Structure Example Structure Example
XO
A. Example 193 Example 201 -\)
A. Example 194 Example 202
Example 195 Example 203
Example 196 Example 204
XX Example 197 Example 205
Example 198 Example 206
Figure imgf000027_0001
XX Example 199 Example 207
CC Example 200 Example 208
\
Figure imgf000028_0001
In another highly preferred embodiment, the compound of the invention is selected from compounds 1-214 above.
A further aspect of the invention relates to a compound as described above for use in medicine.
THERAPEUTIC APPLICATIONS
Another aspect of the invention relates to a compound as described above for use in treating a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
Another aspect relates to the use of a compound as described above in the preparation of a medicament for treating or preventing a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
Preferably, the compound is administered in an amount sufficient to inhibit a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR and IKKepsilon.
Yet another aspect relates to the use of a compound of the invention in the preparation of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity, wherein the kinase is selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, IKKepsilon and combinations thereof.
Preferably, the kinase is selected from TBKl, PDKl, ERK8, MARK3, and IKKepsilon and combinations thereof.
More preferably, the kinase is selected from TBKl and PDKl. Even more preferably, the kinase is TBKl .
In one preferred embodiment, the compound of the invention is exhibits an IC50 value against TBKl of from about 1 μM to about 10 μM, more preferably from about 100 nM to about 1 μM, even more preferably, less than about 100 nM.
Preferably, the disorder is selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, diseases of the eye, including Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, autoimmune diseases, artherosclerosis, retinopathy, osteoarthritis, fibrotic diseases, endometriosis and chronic inflammation.
In the innate immune system TBKl is activated in response to LPS engagement of Toll-like receptor 4 (TLR4) or double-stranded RNA (from double stranded RNA viruses) engagement of TLR3. It is also activated in response to the pro-inflammatory cytokines TNF and IL-I. Once activated TBKl phosphorylates and activates interferon-regulatory factor 3 (IRF3), a transcription factor that triggers the production of interferon beta and chemokines, like interleukin-8 (IL-8) and RANTES. These substances play a key role in mediating host defence against infection by bacteria and viruses. Mice that do not express interferon beta or IRF3 are resistant to LPS-induced septic shock. These observations suggest that a drug that inhibits TBKl may have efficacy for the treatment/prevention of septic shock and/or the treatment of inflammatory disease.
TBKl is also activated in response to hypoxia and stimulates the production of pro-angiogenic factors, such as VEGF and IL-I. The expression of TBKl rises 2.5-3-fold after 24h of hypoxia, similar to the increase in expression of VEGF. The hypoxia induced increase in VEGF expression can be abolished by siRNA "knockdown" of TBKl. The level of TBKl mRNA and protein is elevated in malignant colon and breast cancer cells (see Korherr et al (2006) PNAS 103, 4240-4245 and references therein). TBKl is also recruited and activated by the RalB/Sec5 effector complex; in cancer cells, constitutive engagement of this pathway via chronic RaIB activation, restricts the initiation of apoptotic programmes (Chien et al (2006) Cell 127, 157-170 and references there-in).
For these reasons the drugs that inhibit TBKl may have efficacy for the treatment of cancers.
In one preferred embodiment, the compounds of the invention are useful in the treatment of Primary open Angle Glaucoma (POAG). Primary Open Angle Glaucoma (POAG) is a leading cause of irreversible blindness affecting 35 million people worldwide. The disease is genetically heterogeneous and mutations in the protein optineurin (OPTN) are associated with a form of POAG associated with normal intraocular pressure, termed Normal Tension Glaucoma (NTG) or Low Tension Glaucoma (LTG).1'2 A study of 54 families with autosomal dominant adult onset glaucoma, 17% had one of four sequence mutations in OPTN, the most prevalent being the OPTN[E50K] mutant. How this mutation in OPTN might cause POAG is unknown. However, tumour necrosis factor α (TNFα) has been reported to increase the severity of damage in optic nerve heads of POAG and LTG subjects3'4. Moreover, exposure to TNFα10 induces the de novo expression of optineurin. These observations suggest that some forms of POAG may be caused by an abnormal response to this cytokine.11 The compounds described herein may therefore find use in treating POAG and/or diseases associated with optineurin activity.
Another aspect of the invention relates to a method of treating a TBKl, MKKl, ERK8, RSKl, RSK2, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and/or IKKepsilon and optionally PDKl related disease or disorder. The method according to this aspect of the present invention is effected by administering to a subject in need thereof a therapeutically effective amount of a compound of the present invention, as described hereinabove, either per se, or, more preferably, as a part of a pharmaceutical composition, mixed with, for example, a pharmaceutically acceptable carrier, as is detailed hereinafter.
Yet another aspect of the invention relates to a method of treating a mammal having a disease state alleviated by the inhibition of a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound according to the invention.
Preferably, the disease state is alleviated by the inhibition of TBKl, PDKl, ERK8, MARK3 or IKKepsilon, more preferably TBKl or IKKepsilon, even more preferably TBKl.
Preferably, the mammal is a human. The term "method" refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
The term "administering" as used herein refers to a method for bringing a compound of the present invention and a target kinase together in such a manner that the compound can affect the enzyme activity of the kinase either directly; i.e., by interacting with the kinase itself or indirectly; i.e., by interacting with another molecule on which the catalytic activity of the kinase is dependent. As used herein, administration can be accomplished either in vitro, i.e. in a test tube, or in vivo, i.e., in cells or tissues of a living organism.
Herein, the term "treating" includes abrogating, substantially inhibiting, slowing or reversing the progression of a disease or disorder, substantially ameliorating clinical symptoms of a disease or disorder or substantially preventing the appearance of clinical symptoms of a disease or disorder.
Herein, the term "preventing" refers to a method for barring an organism from acquiring a disorder or disease in the first place.
The term "therapeutically effective amount" refers to that amount of the compound being administered which will relieve to some extent one or more of the symptoms of the disease or disorder being treated.
For any compound used in this invention, a therapeutically effective amount, also referred to herein as a therapeutically effective dose, can be estimated initially from cell culture assays. For example, a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 Or the ICioo as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Initial dosages can also be estimated from in vivo data. Using these initial guidelines one of ordinary skill in the art could determine an effective dosage in humans. Moreover, toxicity and therapeutic efficacy of the compounds described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD50 and the ED50. The dose ratio between toxic and therapeutic effect is the therapeutic index and can be expressed as the ratio between LD50 and ED50. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell cultures assays and animal studies can be used in formulating a dosage range that is not toxic for use in human. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition, (see, e.g., Fingl et al., 1975, In: The Pharmacological Basis of Therapeutics, chapter 1, page 1).
Dosage amount and interval may be adjusted individually to provide plasma levels of the active compound which are sufficient to maintain therapeutic effect. Usual patient dosages for oral administration range from about 50-2000 mg/kg/day, commonly from about 100-1000 mg/kg/day, preferably from about 150-700 mg/kg/day and most preferably from about 250-500 mg/kg/day.
Preferably, therapeutically effective serum levels will be achieved by administering multiple doses each day. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration. One skilled in the art will be able to optimize therapeutically effective local dosages without undue experimentation.
As used herein, kinase related disease or disorder" refers to a disease or disorder characterized by inappropriate kinase activity or over-activity of a kinase as defined herein. Inappropriate activity refers to either; (i) kinase expression in cells which normally do not express said kinase; (ii) increased kinase expression leading to unwanted cell proliferation, differentiation and/or growth; or, (iii) decreased kinase expression leading to unwanted reductions in cell proliferation, differentiation and/or growth. Over-activity of kinase refers to either amplification of the gene encoding a particular kinase or production of a level of kinase activity, which can correlate with a cell proliferation, differentiation and/or growth disorder (that is, as the level of the kinase increases, the severity of one or more of the symptoms of the cellular disorder increases). Over activity can also be the result of ligand independent or constitutive activation as a result of mutations such as deletions of a fragment of a kinase responsible for ligand binding.
Preferred diseases or disorders that the compounds described herein may be useful in preventing, treating and/or studying are cell proliferative disorders, especially cancer such as, but not limited to, papilloma, blastoglioma, Kaposi's sarcoma, melanoma, lung cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, astrocytoma, head cancer, neck cancer, skin cancer, liver cancer, bladder cancer, breast cancer, lung cancer, uterus cancer, prostate cancer, testis carcinoma, colorectal cancer, thyroid cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma, leukemia, lymphoma, Hodgkin's disease and Burkitt's disease.
Another condition to which the compounds described herein may be useful in preventing, treating and/or studying is septic shock.
Another condition to which the compounds described herein may be useful in preventing, treating and/or studying is inflammatory disease.
P. Cohen et al. have observed that TBKl binds in an enhanced manner to the mutant form of optineurin which causes a form of primary open angle glaucoma (POAG).11 The compounds described herein may therefore find use in treating POAG and/or diseases associated with optineurin activity.
A further aspect relates to the use of a compound which is capable of inhibiting the binding of TBKl to a mutant form of OPTN for the manufacture of a medicament for treating POAG and/or a disease where it would be desirable to inhibit or reduce TBKl binding to mutant form of OPTN. One such mutant is the OPTN (E50K) mutant. Suitable compounds may include the compounds identified herein.
In a further aspect there is provided a method of treating a patient suffering from POAG, comprising the step of administering to the subject an effective amount of a compound which is capable of inhibiting an interaction between TBKl and a mutant form of OPTN, associated with POAG. Suitable compounds include those according to Formula I. In a further aspect there is provided a method of treating a patient suffering from a disease associated with abnormal cell proliferation, comprising the step of administering to the subject an effective amount of a compound of the invention.
In a further aspect there is provided a method of treating a patient suffering from septic shock, neurodegenerative diseases, Alzheimer's disease, comprising the step of administering to the subject an effective amount of a compound of the invention.
Thus, the present invention further provides use of compounds as defined herein for the manufacture of medicaments for the treatment of diseases where it is desirable to inhibit TBKl and/or IKK epsilon. Such diseases include colon and breast cancer, septic shock and/or POAG. A number of papers5'6'7 have described that TBKl and IKKepsilon modulate expression of interferon and interferon inducible genes, without affecting induction of pro-inflammatory cytokines. This indicates that the compounds disclosed herein, may find applications in treating/preventing septic shock or viral infection. Mice that do not express interferon beta or IRF3 are resistant to lipopolysaccharide induced septic shock so that inhibitors of TBKl should be expected to have a similar effect.
PHARMACEUTICAL COMPOSTIONS
For use according to the present invention, the compounds or physiologically acceptable salt, ester or other physiologically functional derivative thereof, described herein, may be presented as a pharmaceutical formulation, comprising the compounds or physiologically acceptable salt, ester or other physiologically functional derivative thereof, together with one or more pharmaceutically acceptable carriers therefore and optionally other therapeutic and/or prophylactic ingredients. The carrier(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. The pharmaceutical compositions may be for human or animal usage in human and veterinary medicine.
Examples of such suitable excipients for the various different forms of pharmaceutical compositions described herein may be found in the "Handbook of Pharmaceutical Excipients, 2nd Edition, (1994), Edited by A Wade and PJ Weller. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
Examples of suitable carriers include lactos'e, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like. Examples of suitable diluents include ethanol, glycerol and water.
The choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice. The pharmaceutical compositions may comprise as, or in addition to, the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s), buffer(s), flavouring agent(s), surface active agent(s), thickener(s), preservative(s) (including anti-oxidants) and the like, and substances included for the purpose of rendering the formulation isotonic with the blood of the intended recipient.
Examples of suitable binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and polyethylene glycol.
Examples of suitable lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition. Examples of preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. Antioxidants and suspending agents may be also used.
Pharmaceutical formulations include those suitable for oral, topical (including dermal, buccal and sublingual), rectal or parenteral (including subcutaneous, intradermal, intramuscular and intravenous), nasal and pulmonary administration e.g., by inhalation. The formulation may, where appropriate, be conveniently presented in discrete dosage units and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing into association an active compound with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
Pharmaceutical formulations suitable for oral administration wherein the carrier is a solid are most preferably presented as unit dose formulations such as boluses, capsules or tablets each containing a predetermined amount of active compound. A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine an active compound in a free-flowing form such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, lubricating agent, surface-active agent or dispersing agent. Moulded tablets may be made by moulding an active compound with an inert liquid diluent. Tablets may be optionally coated and, if uncoated, may optionally be scored. Capsules may be prepared by filling an active compound, either alone or in admixture with one or more accessory ingredients, into the capsule shells and then sealing them in the usual manner. Cachets are analogous to capsules wherein an active compound together with any accessory ingredient(s) is sealed in a rice paper envelope. An active compound may also be formulated as dispersible granules, which may for example be suspended in water before administration, or sprinkled on food. The granules may be packaged, e.g., in a sachet. Formulations suitable for oral administration wherein the carrier is a liquid may be presented as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water liquid emulsion.
Formulations for oral administration include controlled release dosage forms, e.g., tablets wherein an active compound is formulated in an appropriate release - controlling matrix, or is coated with a suitable release - controlling film. Such formulations may be particularly convenient for prophylactic use.
Pharmaceutical formulations suitable for rectal administration wherein the carrier is a solid are most preferably presented as unit dose suppositories. Suitable carriers include cocoa butter and other materials commonly used in the art. The suppositories may be conveniently formed by admixture of an active compound with the softened or melted carriers) followed by chilling and shaping in moulds. Pharmaceutical formulations suitable for parenteral administration include sterile solutions or suspensions of an active compound in aqueous or oleaginous vehicles.
Injectible preparations may be adapted for bolus injection or continuous infusion. Such preparations are conveniently presented in unit dose or multi-dose containers which are sealed after introduction of the formulation until required for use. Alternatively, an active compound may be in powder form which is constituted with a suitable vehicle, such as sterile, pyrogen-free water, before use.
An active compound may also be formulated as long-acting depot preparations, which may be administered by intramuscular injection or by implantation, e.g., subcutaneously or intramuscularly. Depot preparations may include, for example, suitable polymeric or hydrophobic materials, or ion- exchange resins. Such long-acting formulations are particularly convenient for prophylactic use. Formulations suitable for pulmonary administration via the buccal cavity are presented such that particles containing an active compound and desirably having a diameter in the range of 0.5 to 7 microns are delivered in the bronchial tree of the recipient. As one possibility such formulations are in the form of finely comminuted powders which may conveniently be presented either in a pierceable capsule, suitably of, for example, gelatin, for use in an inhalation device, or alternatively as a self-propelling formulation comprising an active compound, a suitable liquid or gaseous propellant and optionally other ingredients such as a surfactant and/or a solid diluent. Suitable liquid propellants include propane and the chlorofluorocarbons, and suitable gaseous propellants include carbon dioxide. Self-propelling formulations may also be employed wherein an active compound is dispensed in the form of droplets of solution or suspension.
Such self-propelling formulations are analogous to those known in the art and may be prepared by established procedures. Suitably they are presented in a container provided with either a manually- operable or automatically functioning valve having the desired spray characteristics; advantageously the valve is of a metered type delivering a fixed volume, for example, 25 to 100 microlitres, upon each operation thereof. As a farther possibility an active compound may be in the form of a solution or suspension for use in an atomizer or nebuliser whereby an accelerated airstream or ultrasonic agitation is employed to produce a fine droplet mist for inhalation.
Formulations suitable for nasal administration include preparations generally similar to those described above for pulmonary administration. When dispensed such formulations should desirably have a particle diameter in the range 10 to 200 microns to enable retention in the nasal cavity; this may be achieved by, as appropriate, use of a powder of a suitable particle size or choice of an appropriate valve. Other suitable formulations include coarse powders having a particle diameter in the range 20 to 500 microns, for administration by rapid inhalation through the nasal passage from a container held close up to the nose, and nasal drops comprising 0.2 to 5% w/v of an active compound in aqueous or oily solution or suspension.
Pharmaceutically acceptable carriers are well known to those skilled in the art and include, but are not limited to, 0.1 M and preferably 0.05 M phosphate buffer or 0.8% saline. Additionally, such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, chelating agents, inert gases and the like.
Formulations suitable for topical formulation may be provided for example as gels, creams or ointments. Such preparations may be applied e.g. to a wound or ulcer either directly spread upon the surface of the wound or ulcer or carried on a suitable support such as a bandage, gauze, mesh or the like which may be applied to and over the area to be treated.
Liquid or powder formulations may also be provided which can be sprayed or sprinkled directly onto the site to be treated, e.g. a wound or ulcer. Alternatively, a carrier such as a bandage, gauze, mesh or the like can be sprayed or sprinkle with the formulation and then applied to the site to be treated. According to a further aspect of the invention, there is provided a process for the preparation of a pharmaceutical or veterinary composition as described above, the process comprising bringing the active compound(s) into association with the carrier, for example by admixture.
In general, the formulations are prepared by uniformly and intimately bringing into association the active agent with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product. The invention extends to methods for preparing a pharmaceutical composition comprising bringing a compound of general formula (I) in conjunction or association with a pharmaceutically or veterinarily acceptable carrier or vehicle.
SALTS/ESTERS
The compounds of the invention can be present as salts or esters, in particular pharmaceutically and veterinarily acceptable salts or esters.
Pharmaceutically acceptable salts of the compounds of the invention include suitable acid addition or base salts thereof. A review of suitable pharmaceutical salts may be found in Berge et al, J Pharm Sci, 66, 1-19 (1977). Salts are formed, for example with strong inorganic acids such as mineral acids, e.g. hydrohalic acids such as hydrochloride, hydrobromide and hydroiodide, sulfuric acid, phosphoric acid sulphate, bisulphate, hemisulphate, thiocyanate, persulphate and sulphonic acids; with strong organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Ci-C4)-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluene sulfonic acid. Salts which are not pharmaceutically or veterinarily acceptable may still be valuable as intermediates.
Preferred salts include, for example, acetate, trifluoroacetate, lactate, gluconate, citrate, tartrate, maleate, malate, pantothenate, adipate, alginate, aspartate, benzoate, butyrate, digluconate, cyclopentanate, glucoheptanate, glycerophosphate, oxalate, heptanoate, hexanoate, fumarate, nicotinate, palmoate, pectinate, 3-phenylpropionate, picrate, pivalate, proprionate, tartrate, lactobionate, pivolate, camphorate, undecanoate and succinate, organic sulphonic acids such as methanesulphonate, ethanesulphonate, 2-hydroxyethane sulphonate, camphorsulphonate, 2- naphthalenesulphonate, benzenesulphonate, p-chlorobenzenesulphonate and p-toluenesulphonate; and inorganic acids such as hydrochloride, hydrobromide, hydroiodide, sulphate, bisulphate, hemisulphate, thiocyanate, persulphate, phosphoric and sulphonic acids.
Esters are formed either using organic acids or alcohols/hydroxides, depending on the functional group being esterified. Organic acids include carboxylic acids, such as alkanecarboxylic acids of 1 to 12 carbon atoms which are unsubstituted or substituted (e.g., by halogen), such as acetic acid; with saturated or unsaturated dicarboxylic acid, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or tetraphthalic; with hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid; with aminoacids, for example aspartic or glutamic acid; with benzoic acid; or with organic sulfonic acids, such as (Ci-C4)-alkyl- or aryl-sulfonic acids which are unsubstituted or substituted (for example, by a halogen) such as methane- or p-toluene sulfonic acid. Suitable hydroxides include inorganic hydroxides, such as sodium hydroxide, potassium hydroxide, calcium hydroxide, aluminium hydroxide. Alcohols include alkanealcohols of 1-12 carbon atoms which may be unsubstituted or substituted, e.g. by a halogen).
ENANTIOMERS/TAUTOMERS
In all aspects of the present invention previously discussed, the invention includes, where appropriate all enantiomers, diastereoisomers and tautomers of the compounds of the invention. The person skilled in the art will recognise compounds that possess optical properties (one or more chiral carbon atoms) or tautomeric characteristics. The corresponding enantiomers and/or tautomers may be isolated/prepared by methods known in the art. Enantiomers are characterised by the absolute configuration of their chiral centres and described by the R- and S-sequencing rules of Cahn, Ingold and Prelog. Such conventions are well known in the art (e.g. see 'Advanced Organic Chemistry', 3rd edition, ed. March, J., John Wiley and Sons, New York, 1985). Compounds of the invention containing a chiral centre may be used as a racemic mixture, an enantiomerically enriched mixture, or the racemic mixture may be separated using well-known techniques and an individual enantiomer may be used alone.
STEREO AND GEOMETRIC ISOMERS Some of the compounds of the invention may exist as stereoisomers and/or geometric isomers - e.g. they may possess one or more asymmetric and/or geometric centres and so may exist in two or more stereoisomeric and/or geometric forms. The present invention contemplates the use of all the individual stereoisomers and geometric isomers of those inhibitor agents, and mixtures thereof. The terms used in the claims encompass these forms, provided said forms retain the appropriate functional activity (though not necessarily to the same degree).
The present invention also includes all suitable isotopic variations of the agent or a pharmaceutically acceptable salt thereof. An isotopic variation of an agent of the present invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that can be incorporated into the agent and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine such as 2H, 3H, 13C, 14C, 15N, 17O, 18O, 31P, 32P, 35S, 18F and 36Cl, respectively. Certain isotopic variations of the agent and pharmaceutically acceptable salts thereof, for example, those in which a radioactive isotope such as 3H or 14C is incorporated, are useful in drug and/or substrate tissue distribution studies. Tritiated, i.e., 3H, and carbon- 14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with isotopes such as deuterium, i.e., 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements and hence may be preferred in some circumstances. For example, the invention includes compounds of general formula (I) where any hydrogen atom has been replaced by a deuterium atom. Isotopic variations of the agent of the present invention and pharmaceutically acceptable salts thereof of this invention can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
PRODRUGS The invention further includes the compounds of the present invention in prodrug form, i.e. covalently bonded compounds which release the active parent drug according to general formula (I) in vivo. Such prodrugs are generally compounds of the invention wherein one or more appropriate groups have been modified such that the modification may be reversed upon administration to a human or mammalian subject. Reversion is usually performed by an enzyme naturally present in such subject, though it is possible for a second agent to be administered together with such a prodrug in order to perform the reversion in vivo. Examples of such modifications include ester (for example, any of those described above), wherein the reversion may be carried out be an esterase etc. Other such systems will be well known to those skilled in the art.
SOLVATES
The present invention also includes solvate forms of the compounds of the present invention. The terms used in the claims encompass these forms.
POLYMORPHS
The invention further relates to the compounds of the present invention in their various crystalline forms, polymorphic forms and (an)hydrous forms. It is well established within the pharmaceutical industry that chemical compounds may be isolated in any of such forms by slightly varying the method of purification and or isolation form the solvents used in the synthetic preparation of such compounds.
ADMINISTRATION
The pharmaceutical compositions of the present invention may be adapted for rectal, nasal, intrabronchial, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intraarterial and intradermal), intraperitoneal or intrathecal administration. Preferably the formulation is an orally administered formulation. The formulations may conveniently be presented in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose. By way of example, the formulations may be in the form of tablets and sustained release capsules, and may be prepared by any method well known in the art of pharmacy. Formulations for oral administration in the present invention may be presented as: discrete units such as capsules, gellules, drops, cachets, pills or tablets each containing a predetermined amount of the active agent; as a powder or granules; as a solution, emulsion or a suspension of the active agent in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water- in-oil liquid emulsion; or as a bolus etc. Preferably, these compositions contain from 1 to 250 mg and more preferably from 10-100 mg, of active ingredient per dose.
For compositions for oral administration (e.g. tablets and capsules), the term "acceptable carrier" includes vehicles such as common excipients e.g. binding agents, for example syrup, acacia, gelatin, sorbitol, tragacanth, polyvinylpyrrolidone (Povidone), methylcellulose, ethylcellulose, sodium carboxymethylcellulose, hydroxypropylmethylcellulose, sucrose and starch; fillers and carriers, for example corn starch, gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid; and lubricants such as magnesium stearate, sodium stearate and other metallic stearates, glycerol stearate stearic acid, silicone fluid, talc waxes, oils and colloidal silica. Flavouring agents such as peppermint, oil of wintergreen, cherry flavouring and the like can also be used. It may be desirable to add a colouring agent to make the dosage form readily identifiable. Tablets may also be coated by methods well known in the art.
A tablet may be made by compression or moulding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active agent in a free flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface-active or dispersing agent. Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may be optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active agent.
Other formulations suitable for oral administration include lozenges comprising the active agent in a flavoured base, usually sucrose and acacia or tragacanth; pastilles comprising the active agent in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active agent in a suitable liquid carrier. Other forms of administration comprise solutions or emulsions which may be injected intravenously, intraarterially, intrathecally, subcutaneously, intradermally, intraperitoneally or intramuscularly, and which are prepared from sterile or sterilisable solutions. Injectable forms typically contain between 10 - 1000 mg, preferably between 10 - 250 mg, of active ingredient per dose.
The pharmaceutical compositions of the present invention may also be in form of suppositories, pessaries, suspensions, emulsions, lotions, ointments, creams, gels, sprays, solutions or dusting powders. An alternative means of transdermal administration is by use of a skin patch. For example, the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin. The active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
DOSAGE
A person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation. Typically, a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy. The dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
In accordance with this invention, an effective amount of a compound of general formula (I) may be administered to inhibit the kinase implicated with a particular condition or disease. Of course, this dosage amount will further be modified according to the type of administration of the compound. For example, to achieve an "effective amount" for acute therapy, parenteral administration of a compound of general formula (I) is preferred. An intravenous infusion of the compound in 5% dextrose in water or normal saline, or a similar formulation with suitable excipients, is most effective, although an intramuscular bolus injection is also useful. Typically, the parenteral dose will be about 0.01 to about 100 mg/kg; preferably between 0.1 and 20 mg/kg, in a manner to maintain the concentration of drug in the plasma at a concentration effective to inhibit a kinase. The compounds may be administered one to four times daily at a level to achieve a total daily dose of about 0.4 to about 400 mg/kg/day. The precise amount of an inventive compound which is therapeutically effective, and the route by which such compound is best administered, is readily determined by one of ordinary skill in the art by comparing the blood level of the agent to the concentration required to have a therapeutic effect.
The compounds of this invention may also be administered orally to the patient, in a manner such that the concentration of drug is sufficient to achieve one or more of the therapeutic indications disclosed herein. Typically, a pharmaceutical composition containing the compound is administered at an oral dose of between about 0.1 to about 50 mg/kg in a manner consistent with the condition of the patient. Preferably the oral dose would be about 0.5 to about 20 mg/kg.
No unacceptable toxicological effects are expected when compounds of the present invention are administered in accordance with the present invention. The compounds of this invention, which may have good bioavailability, may be tested in one of several biological assays to determine the concentration of a compound which is required to have a given pharmacological effect.
COMBINATIONS
In a particularly preferred embodiment, the one or more compounds of the invention are administered in combination with one or more other active agents, for example, existing drugs available on the market. In such cases, the compounds of the invention may be administered consecutively, simultaneously or sequentially with the one or more other active agents.
Drugs in general are more effective when used in combination. In particular, combination therapy is desirable in order to avoid an overlap of major toxicities, mechanism of action and resistance mechanism(s). Furthermore, it is also desirable to administer most drugs at their maximum tolerated doses with minimum time intervals between such doses. The major advantages of combining chemotherapeutic drugs are that it may promote additive or possible synergistic effects through biochemical interactions and also may decrease the emergence of resistance. Beneficial combinations may be suggested by studying the inhibitory activity of the test compounds with agents known or suspected of being valuable in the treatment of a particular disorder. This procedure can also be used to determine the order of administration of the agents, i.e. before, simultaneously, or after delivery. Such scheduling may be a feature of all the active agents identified herein.
ASSAY
A further aspect of the invention relates to the use of a compound as described above in an assay for identifying further candidate compounds capable of inhibiting one or more kinases selected from TBK 1 , MKK 1 , ERO, RSK 1 , RSK2, PDK 1 , S6K 1 , MNK2, PHK, CHK 1 , CHK2, GSObeta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon.
Preferably, the assay is a competitive binding assay.
More preferably, the competitive binding assay comprises contacting a compound of the invention with a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, and a candidate compound and detecting any change in the interaction between the compound according to the invention and the kinase.
Preferably, the candidate compound is generated by conventional SAR modification of a compound of the invention.
As used herein, the term "conventional SAR modification" refers to standard methods known in the art for varying a given compound by way of chemical derivatisation.
Thus, in one aspect, the identified compound may act as a model (for example, a template) for the development of other compounds. The compounds employed in such a test may be free in solution, affixed to a solid support, borne on a cell surface, or located intracellularly. The abolition of activity or the formation of binding complexes between the compound and the agent being tested may be measured. The assay of the present invention may be a screen, whereby a number of agents are tested. In one aspect, the assay method of the present invention is a high through-put screen.
This invention also contemplates the use of competitive drug screening assays in which neutralising antibodies capable of binding a compound specifically compete with a test compound for binding to a compound.
Another technique for screening provides for high throughput screening (HTS) of agents having suitable binding affinity to the substances and is based upon the method described in detail in WO 84/03564.
It is expected that the assay methods of the present invention will be suitable for both small and large-scale screening of test compounds as well as in quantitative assays.
Preferably, the competitive binding assay comprises contacting a compound of the invention with a kinase in the presence of a known substrate of said kinase and detecting any change in the interaction between said kinase and said known substrate.
A further aspect of the invention provides a method of detecting the binding of a ligand to a kinase, said method comprising the steps of:
(i) contacting a ligand with a kinase in the presence of a known substrate of said kinase;
(ii) detecting any change in the interaction between said kinase and said known substrate; and wherein said ligand is a compound of the invention.
One aspect of the invention relates to a process comprising the steps of:
(a) performing an assay method described hereinabove;
(b) identifying one or more ligands capable of binding to a ligand binding domain; and
(c) preparing a quantity of said one or more ligands. Another aspect of the invention provides a process comprising the steps of:
(a) performing an assay method described hereinabove;
(b) identifying one or more ligands capable of binding to a ligand binding domain; and
(c) preparing a pharmaceutical composition comprising said one or more ligands.
Another aspect of the invention provides a process comprising the steps of:
(a) performing an assay method described hereinabove;
(b) identifying one or more ligands capable of binding to a ligand binding domain;
(c) modifying said one or more ligands capable of binding to a ligand binding domain; (d) performing the assay method described hereinabove;
(e) optionally preparing a pharmaceutical composition comprising said one or more ligands.
The invention also relates to a ligand identified by the method described hereinabove.
Yet another aspect of the invention relates to a pharmaceutical composition comprising a ligand identified by the method described hereinabove.
Another aspect of the invention relates to the use of a ligand identified by the method described hereinabove in the preparation of a pharmaceutical composition for use in the treatment of one or more disorders selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, diseases of the eye, including Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, autoimmune diseases, artherosclerosis, retinopathy, osteoarthritis, fϊbrotic diseases, endometriosis and chronic inflammation.
The above methods may be used to screen for a ligand useful as an inhibitor of one or more kinases.
Compounds of general formula (I) are useful both as laboratory tools and as therapeutic agents. In the laboratory certain compounds of the invention are useful in establishing whether a known or newly discovered kinase contributes a critical or at least significant biochemical function during the establishment or progression of a disease state, a process commonly referred to as 'target validation'. SYNTHESIS
A further aspect of the invention relates to a process for preparing a compound as described above, said process comprising the steps of:
Figure imgf000050_0001
Ia II III I
(i) converting a compound of formula Ia to a compound of formula II, where each LG is independently a leaving group; (ii) reacting said compound of formula II with an amine of formula IV to form a compound of formula III; (iii) reacting said compound of formula III with an amine of formula VII to form a compound of formula I.
Scheme 1 illustrates the conversion of compounds of formula (Ia) to compounds of formula (II), wherein Rl, R3 and R7 are as defined previously.
Scheme 1
Figure imgf000050_0002
Ia II HI
Compounds of formula (Ia) can be obtained from commercial sources or by following known literature procedures; Rl, R3 and R7 are as defined previously; LG refers to a leaving group such as halogen, methoxy, thiomethyl, triflate, tosylate or the group -SO2Me (but is preferably chlorine).
Compounds with formula (IV) are commercially available, known in the literature or are readily obtainable by the skilled person by following standard chemical procedures.
Step (a)
Compounds of formula (Ia) can be converted to compounds of formula (II) by treatment with POCl3 in the presence of an additive, such as N,N-diisopropylethylamine at temperatures in the range of 5O0C to reflux for reaction times up to 24 h.
Preferred conditions: leq. of formula 1, leq of N,N-diisopropylethylamine, 7.8eq. of POCl3 heating at 90 0C for 2 h.
Step (b)
This step involves the displacement of a leaving group (LG), preferably chlorine, in formula (II) with an amino group of formula (IV) in a suitable solvent (such as isopropanol or dioxane) in the presence of an organic base (such as N,N-diisopropylethylamine) at temperatures in the range of 0- 80 0C for reaction times of up to 24 h.
Preferred conditions: leq. of formula (II), leq. of formula (IV), 3eq. ofΝ-Ν-diisopropylethylamine in isopropanol at room temperature overnight.
Scheme 2
Figure imgf000051_0001
Scheme 2 illustrates the conversion of compounds with formula (V) to compounds with formula (VI), wherein Rl, R2, R3, R7 and LG are as defined previously. Compounds of formula (V) can be converted to compounds with formula (VI) by displacement of a leaving group (preferably chlorine) in formula (V) with an amine with formula (VII) in an appropriate solvent (e.g. acetonitrile or n-butanol) in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H2SO4, p-toluenesulfonic acid). The reaction can be carried out at temperatures ranging from 50 0C to 200 0C by convectional heating or microwave heating. Preferred method: leq. of formula (V), leq. of acetic acid, 3eq. of formula (VII) in n-butanol heated to 150 0C in the microwave for 40 minutes.
Step fd^ Alternatively, compounds of formula (VI) can be prepared from compounds with formula (V) (where LG is chlorine) by reaction with amine (VII) in the presence of a palladium source (e.g. Pd(OAc)2 or Pd2(dba)3), a suitable ligand (e.g. bis(diphenylphosphino)-9,9-dimethylxanthene) and a suitable base (e.g. Cs2CO3 or sodium tert-butoxide) in a suitable solvent (e.g. dioxane). The reaction is generally carried out at around the reflux temperature of the solvent under an inert atmosphere.
Preferred method: leq. of formula (V), 1.2eq. of amine (VII), 0.06eq. of Pd2(dba)3, 0.12eq. of bis(diphenylphosphino)-9,9-dimethylxanthene, 3eq. of sodium tert-butoxide in dioxane at 105 0C for 18 h.
Scheme 3
Figure imgf000052_0001
(0
Compounds of formula (IX) and formula (X) can be prepared from compounds of formula (VIII) according to scheme 3, wherein Rl, R2, R7 and LG are as defined previously; Q is NR7 or oxygen and PG represents a suitable protecting group (such as tert-butoxycarbonyl or benzyloxycarbonyl). Compounds of formula (VIII) wherein Q = NH, can be prepared according to step (b) Scheme 1 ; compounds of formula (VIII) wherein Q = oxygen can be prepared according to Scheme 7. Compounds of formula (IX) and formula (X) can be prepared from compounds of formula (VIII) by displacement of a leaving group (preferably chlorine) in formula (VIII) with an amine with formula (VII) in an appropriate solvent (e.g. acetonitrile or n-butanol) in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H2SO4, p-toluenesulfonic acid). The reaction can be carried out at temperatures typically ranging from 50 0C to 200 0C, either by convectional heating or microwave heating. It will be understood by a skilled person that certain protecting groups are sensitive to the reaction conditions and may therefore give the product in the unprotected form (formula X), or indeed give mixtures of both formula (IX) and formula (X). Preferred conditions: leq of formula (VIII), 1.3 eq of formula (VII), 1.45eq of 4M HCl in dioxane, in aqueous acetonitrile at 50 0C overnight.
Step (f)
Compounds of formula (X) can be prepared from compounds of formula (IX) using standard methods for amine deprotection known to a skilled person. For example, where PG is tert- butoxycarbonyl reaction of formula (IX) with a suitable acid (such as HCl or trifluoroacetic acid) in a suitable solvent (such as dioxane or DCM) at temperatures typically in the range of 0 0C to the reflux temperature of the solvent enables this transformation to occur. Preferred conditions: Formula (IX) is stirred in 4M HCl in dioxane at room temperature for 2 h.
Scheme 4
Figure imgf000053_0001
Compounds with formula (XI) can be prepared according to Scheme 4, wherein Rl, R2, R6, R7 and Q are as defined previously, X refers to either OH or chlorine. Compounds with formula (XI) can be prepared from compounds of formula (X) by treatment with formula (XII), where X is chlorine. The reaction is carried out in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine) in a suitable solvent (such as DCM or dioxane) at temperatures ranging from O0C to the reflux temperature of the solvent Preferred conditions: leq of formula (X), 1.02eq of formula (XII), 2.2eq of triethylamine in DCM at room temperature for 2 h.
Step (h)
Alternatively, compounds with formula (XI) can be prepared from compounds of formula (X) by treatment with formula (XII), where X is hydroxy. The reaction is carried out in the presence of a suitable amine coupling reagent known to the skilled person, (such as but not limited to HATU, DCC, EDCI), optionally in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine), optionally in the presence of an additive (such as but not limited to, 1- hydroxybenzotriazole) in a suitable solvent (such as DMF or pyridine) at temperatures ranging from 00C to the reflux temperature of the solvent.
Preferred conditions: leq of formula (X), l.όeq of formula (XII), 1.7eq of HATU, 6.2eq of N,N- diisopropylethylamine in DMF at room temperature overnight.
Scheme 5
Figure imgf000054_0001
Alternatively, compounds with formula (XI) can be prepared according to Scheme 5 wherein Rl, R2, R6 and R7, LG are all as defined previously; X is chlorine or hydroxyl. Compounds with formula (VIII), wherein PG is tert-butoxycarbonyl and LG is preferably chlorine are converted to formula (XIII) on treatment with a suitable acid (such as HCl or trifluoroacetic acid) in a suitable solvent (such as dioxane or DCM) at temperatures typically in the range of 0 0C to the reflux temperature of the solvent. Typical reaction times are in the range of 1 h to 24 h. Preferred conditions: Formula (VIII) is stirred in 4M HCl in dioxane at room temperature for 1 h.
Step
Compounds with formula (XIV) can be prepared from compounds of formula (XIII) by treatment with formula (XII), wherein X is chlorine. The reaction is carried out in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine) in a suitable solvent (such as DCM or dioxane) at temperatures ranging from 00C to the reflux temperature of the solvent.
Preferred conditions: leq of formula (XIII), 1.02eq of formula (XII), 2.2eq of triethylamine in
DCM at room temperature for 2 h.
Step (k)
Alternatively, compounds with formula (XIV) can be prepared from compounds of formula (XIII) by treatment with formula (XII), wherein X is hydroxyl. The reaction is carried out in the presence of a suitable amine coupling reagent known to the skilled person, (such as but not limited to HATU, DCC, EDCI), optionally in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine), optionally in the presence of an additive (such as but not limited to, hydroxybenzotriazole) in a suitable solvent (such as DMF or pyridine) at temperatures ranging from O0C to the reflux temperature of the solvent.
Preferred conditions: leq of formula (XIII), 1.6eq of formula (XII), 1.7eq of HATU, 6.2eq of N1N- diisopropylethylamine in DMF at room temperature overnight.
Step (T)
Compounds of formula (XIV) can be converted to compounds with formula (XI) by displacement of a leaving group (preferably chlorine) in formula (XIV) with an amine with formula (VII) in an appropriate solvent (e.g. acetonitrile or n-butanol) in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H2SO4, p-toluenesulfonic acid). The reaction can be carried out at temperatures typically ranging from 50 0C to 200 0C, by either convectional heating or optionally with microwave heating.
Preferred method: leq. of formula (XIV), leq. of acetic acid, 3eq. of formula (VII) in n-butanol heated to 150 0C in the microwave for 40 minutes.
Step (m)
Alternatively, compounds of formula (XI) can be prepared from compounds with formula (XIV)
(wherein LG is chlorine) by reaction with amine (VII) in the presence of a palladium source (e.g.
Pd(OAc)2 or Pd2(dba)3), a suitable ligand (e.g. bis(diphenylphosphino)-9,9-dimethylxanthene) and a suitable base (e.g. Cs2CO3 or sodium terf-butoxide) in a suitable solvent (e.g. dioxane). The reaction is generally carried out at around the reflux temperature of the solvent under an inert atmosphere.
Preferred method: leq. of formula (XIV), 1.2eq. of amine (VII), 0.06eq. of Pd2(dba)3, 0.12eq. of bis(diphenylphosphino)-9,9-dimethylxanthene, 3eq. of sodium tert-butoxide in dioxane at 105 0C for 18 h.
Scheme 6
Figure imgf000056_0001
Compounds with formula (XVI) can be prepared from compounds of formula (XV) according to Scheme 6, wherein Rl, R2, R4, R5 and R6 are as defined previously. Compounds of formula (XV) can be prepared according to step (c), scheme 2.
Step (n)
Compounds of with formula (XVI) can be prepared from compounds of formula (XV) by treatment with a suitable amine with the formula NHR4R5. The reaction is carried out in the presence of a suitable amine coupling reagent known to the skilled person, (such as but not limited to HATU, DCC, EDCI), optionally in the presence of a suitable base (such as N,N-diisopropylethylamine or triethylamine), optionally in the presence of an additive (such as but not limited to, 1- hydroxybenzotriazole) in a suitable solvent (such as DMF or pyridine) at temperatures ranging from 00C to the reflux temperature of the solvent.
Preferred conditions: leq of formula (XV), 1.5eq of amine, 1.6eq of HATU, 6eq of
N,N-diisopropylethylamine in DMF at room temperature overnight.
A further aspect of the invention relates to a process for preparing a compound of formula XIX, wherein R1, R2 and R3 are as defined above, said process comprising the steps of:
Figure imgf000057_0001
(X=O, S)
Ia II XVIII XIX
(i) converting a compound of formula Ia to a compound of formula II, where each LG is independently a leaving group; (ii) reacting said compound of formula II with an alkohol of formula XVII to form a compound of formula XVIII; (iii) reacting said compound of formula XVIII with an amine of formula VII to form a compound of formula XIX.
Compounds with formula (XIX) can be prepared according to scheme 7, wherein LG, Rl, R2 and R3 are as defined previously. Scheme 7
Figure imgf000057_0002
Compounds with formula XVIII can be prepared by reacting compounds with formula (XVII) with compounds with formula (II) in a suitable solvent (such as THF) in the presence of a strong base, such as sodium hydride at temperatures ranging from 0 0C to the reflux temperature of the solvent. Preferred conditions: leq. of formula (XVII) is deprotonated with leq. of sodium hydride in THF at O0C, followed by treatment with leq. of formula (II) at room temperature overnight.
Step ( pi
Compounds of formula (XVIII) can be converted to compounds with formula (XIX) by displacement of a leaving group (preferably chlorine) in formula (XVIII) with an amine with formula (VII) in an appropriate solvent (e.g. n-butanol) in the presence of a suitable organic acid or mineral acid (such as acetic acid, HCl, H2SO4, p-toluenesulfonic acid). The reaction can be carried out at temperatures typically ranging from 50 0C to 200 0C, by either convectional heating or optionally with microwave heating. Preferred method: leq. of formula (XVIII), 0.2eq. of acetic acid, 2eq. of formula (IV) in n-butanol heated to 150 0C in the microwave for 15 minutes.
The present invention is further described by way of the following non-limiting examples.
EXAMPLES
Materials and Methods
Source and purification of kinases
All protein kinases were of human origin and encoded full length proteins, unless indicated otherwise. They were either expressed as glutathione S-transferase (GST) fusion proteins in Escherichia CoIi or as hexahistidine (Hisό)-tagged proteins in insect Sf21 cells. GST fusion proteins were purified by affinity chromatography on glutathione- Sepharose, and His6-tagged proteins on nickel/nitriloacetate-agarose. The procedures for expressing some of the protein kinases used herein have been detailed previously.8'9 The following sections outline the DNA vectors synthesised and the procedures used to express and purifying protein kinases that have not been reported previously. Expression in E.coli
The following proteins were expressed in E.coli:- CHK2[5-543], cyclin-dependent protein kinase 2 (CDK2), MAP kinase-interacting kinase 2 (MNK2), extra-cellular signal-regulated kinase 1 (ERKl).
Expression in Sf21 cells
The following kinases were expressed in Sf21 cells:Aurora B and Aurora C, extra-cellular signal- regulated kinase 8 (ERK8), microtubule affinity regulating kinase 3 (MARK3), protein kinase Bα[118-480][S473D], protein kinase Bβ (PKBβ[120-481][S474D], 3-phosphoinositide-dependent protein kinase- 1 [52-556]
(PDKl [52-556], IKKε, TBKl,
Activation of protein kinases
In order to produce activated forms of Aurora B and Aurora C, insect Sf21 cells were incubated for I h with the protein phosphatase inhibitor okadaic acid (50 nM). JNK isoforms were activated with
MKK4 and MKK7, MNK2 with p38α MAPkinase; PKBα, PKBβ, with PDKl, and ERKl with
MKKl. To activate CDK2, bacterial pellets expressing cyclin A2 and CDK2 were mixed together, lyse and purified on glutathione Sepharose. The GST-tags were removed by cleavage with
PreScission protease and the CDK2-cyclin A2 complex purified by chromatography on SP- Sepharose. It was then activated with CAK1/CDK7 followed by chromatography on nickel- nitrilotriacetate agarose to remove CAK1/CDK7, which binds to this column by virtue of its C- terminal His6 tag. All the other protein kinases were active as expressed.
Protein kinase assays All assays (25.5μl) were carried out at room temperature (21°C) and were linear with respect to time and enzyme concentration under the conditions used. Assays were performed for 30 min using Multidrop Micro reagent dispensers (Thermo Electron Corporation, Waltham, MA 02454, USA) in a 96- well format. The concentration of magnesium acetate in the assays was 10 mM and the [γ- 33P] ATP (800 cpm / pmol) was used at 5, 20 or 50 μM as indicated, in order to be at or below the Km for ATP for each enzyme. Protein kinases assayed at 5 μM ATP were:- ERKl, ERK8, PKBα, MARK3, Aurora C. Protein kinases assayed at 20 μM ATP were:- JNKl, PDKl5CHKl, CHK2, CDK2 and Aurora B. Protein kinases assayed at 50 μM ATP were:-MNK2, IKKepsilon and TBKl . The assays were initiated with MgATP, stopped by addition of 5 μl of 0.5 M orthophosphoric acid and spotted on to P81 filterplates using a unifilter harvester (PerkinElmer, Boston, MA 02118, USA). The IC50 values of inhibitors were determined after carrying out assays at 10 different concentrations of each compound.
ERKl and ERK8 were both assayed against myelin basic protein (MBP, 0.33mg/ml). MARK3 was assayed against the peptide KKKVSRSGL YRSPSMPENLNRPR (300μM), MNK2 against the eIF4E protein (0.5mg/ml). PKBβ was assayed against the peptide GRPRTSSFAEGKK (30μM). TBKl were assayed against (AKPKGNKD YHLQTCCGSLA YRRR) (300 μM). The substrates used for other protein kinases were described previously.8'9
Unless stated otherwise, enzymes were diluted in 50 mM Tris/HCl pH 7.5, 0.1 mM EGTA, 1 mg/ml BSA, 0.1% (v/v) 2-mercaptoethanol and assayed in 50 mM Tris/HCl pH 7.5, 0.1 mM EGTA, 0.1 % (v/v) 2-mercaptoethanol.
General procedures for synthesis of compounds Chromatography
Preparative high pressure liquid chromatography was carried out using apparatus made by Agilent. The apparatus is constructed such that the chromatography (column: either a 30 x 100 mm (10 μm) C-18 Phenomenex Gemini column at a flow rate of 50 ml/min, or a 21.2 x 100 mm (5 μm) C-18 Phenomenex Gemini column at a flow rate of 20 ml/min) is monitored by a multi-wavelength UV detector (G1365B manufactured by Agilent) and an MM-ES+APCI mass spectrometer (G-1956A, manufactured by Agilent) connected in series, and if the appropriate criteria are met the sample is collected by an automated fraction collector (G1364B manufactured by Agilent). Collection can be triggered by any combination of UV or mass spectrometry or can be based on time. Typical conditions for the separation process are as follows: The gradient is run over a 10 minute period (gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: either 0.1% trifluoroacetic acid is added to the water (low pH buffer), or ammonium bicarbonate (10 mmol / 1) and 35% ammonium hydroxide (1.6 ml / 1) is added to the water (high pH buffer). It will be appreciated by those skilled in the art that it may be necessary or desirable to modify the conditions for each specific compound, for example by changing the solvent composition at the start or at the end, modifying the solvents or buffers, changing the run time or changing the flow rate.
Flash chromatography refers to silica gel chromatography and carried out using an SP4 MPLC system (manufactured by Biotage); pre-packed silica gel cartridges (supplied by Biotage); or using conventional glass column chromatography.
Analytical Methods
1H Nuclear magnetic resonance (NMR) spectroscopy was carried out using an ECX400 spectrometer (manufactured by JEOL) in the stated solvent at around room temperature unless otherwise stated. In all cases, NMR data were consistent with the proposed structures. Characteristic chemical shifts (δ) are given in parts-per-million using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; dd, doublet of doublets; br, broad. Mass spectra were recorded using a MM-ES+APCI mass spectrometer (G- 1956A, manufactured by Agilent). Where thin layer chromatography (TLC) has been used it refers to silica gel TLC using silica gel MK6F 6θA plates, Rf is the distance travelled by the compound divided by the distance travelled by the solvent on a TLC plate. Compound preparation Where the preparation of starting materials is not described, these are commercially available, known in the literature, or readily obtainable by those skilled in the art using standard procedures. Where it is stated that compounds were prepared analogously to earlier or later examples or intermediates, it will be appreciated by the skilled person that the reaction time, number of equivalents of reagents and temperature can be modified for each specific reaction and that it may be necessary or desirable to employ different work-up or purification techniques. Where reactions are carried out using microwave irradiation, the microwave used is an Initiator 60 supplied by Biotage. The actual power supplied varies during the course of the reaction in order to maintain a constant temperature.
Abbreviations DCM = Dichloromethane
DMF = N,N-Dimethylformamide
THF = Tetrahydrofuran MeOH = Methanol
TFA = Trifluoroacetic acid
Xantphos = 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene
HATU =N,N,Nl,N'-Tetramethyl-0-(7-azabenzotriazol- 1 -yl)uronium- hexafluorophospate EDCI = 1,3-Propanediamine, N3-(ethylcarbonimidoyl)-Nl, Nl -dimethyl-, hydrochloride
DCC = l^-Dicyclohexylcarbodiimide
Pd2(dba)3 = tτis(dibenzylideneacetone)dipalladium(0)
TEA = Triethylamine rm = Reaction mixture rt = Room temperature
AcOH = Acetic acid
IPA = Isopropanol
DIPEA = N,N-diisopropylethylamine
TBSMSCl = Tertiarybutyldimethylsilyl chloride MeCN = Acetonitrile
NH3 = Ammonia
EtOH = Ethanol
EtOAc = Ethyl Acetate
LCMS = Mass spectrometry directed high pressure liquid chromatography UV = Ultraviolet
SCX = Strong cation exchange
Examples
Intermediate 1 2, ^DichloroS-cyclopropyl-pyrimidine
Figure imgf000062_0001
To a suspension of 5-cyclopropyl-2-thioxo-2,3-dihydro-l//-pyrimidin-4-one [Prepared according to J. Org. Chem. 1994, 4791-4799] (2.3 g, 13.7 mmol) in phosphorus oxychloride (10 ml, 107 mmol) was added N,N-diisopropylethylamine (3.5 ml, 13.8 mmol) dropwise at O0C. The resulting mixture was stirred at 90 0C for 2 h to give complete reaction. The mixture was then concentrated to dryness, diluted with DCM (50 ml) and poured into ice water (50 ml). The organic layer was separated, the aqueous phase was extracted with DCM (x2) and the combined organic phases were dried (MgSO4) and evaporated to dryness. The crude yellow oil was purified by flash chromatography on the Biotage SP4, eluting with 0 to 30% ethyl acetate/petroleum ether gradient to give a pale yellow oil which solidified on standing to an off-white solid (2.32 g, 90 % yield). 1H NMR (400 MHz, DMSCW6) δ ppm 0.91 (m, 2 H), 1.07 (m, 2 H), 1.97 - 2.05 (m, 1 H), 8.49 (s, 1 H). Rf (10% ethyl acetate in petroleum ether) = 0.75.
Intermediate 2
[3-(2-Chloro-5-cyclopropyl-pyrimidin-4-ylamino)-propyl]-carbamic acid tert-butyl ester
Figure imgf000063_0001
Intermediate 1 (5.84 g, 30.9 mmol), 3-aminopropyl)carbamic acid tert-buty\ ester (5.38 g, 30.9 mmol) and N,N-diisopropylethylamine (16.5 ml, 92.7 mmol) were combined in isopropyl alcohol (150 ml) at 0 °C and then stirred at this temperature for 1 hour. After this time, the reaction mixture was warmed up to room temperature and stirring was continued overnight. The volatiles were evaporated and the crude material dissolved in ethyl acetate, washed with water, followed by brine, dried (MgSO4) and solvents evaporated to dryness. The crude product was purified by flash chromatography using a Biotage SP4 (ethyl acetate / petroleum ether gradient) to give the product as a white solid (7.1 g, 70%). 1H ΝMR (400 MHz, DMSO-<i6) δ ppm 0.53 - 0.59 (m, 2 H), 0.84 - 0.92 (m, 2 H), 1.37 (s, 9 H), 1.47 - 1.55 (m, 1 H), 1.60 - 1.69 (m, 2 H), 2.93 - 3.02 (m, 2 H), 3.33 - 3.41 (m, 2 H), 6.87 (t, J=5.7 Hz, 1 H), 7.41 (t, J=5.7 Hz, 1 H), 7.69 (s, 1 H); m/z (ES+APCI)+ 327 [M+H]+.
Intermediate 3
N4-(3-Amino-propyl)-5-cyclopropyl-N2-(3-fluoro-phenyl)-pyrimidine-2,4-diamine hydrochloride
Figure imgf000064_0001
To a solution of Intermediate 2 (2 g, 6.1 mmol) and 3-fluoroaniline (766 μl, 8.0 mmol) in 14:1 acetonitrile : water (53.6 ml) was added 4M HCl in dioxane (2.22 ml, 8.9 mmol). The reaction mixture was heated to 50 0C and stirred at this temperature overnight. The white precipitate that formed was filtered, washed with diethyl ether and dried under vacuum to give a white solid (1.3 g, 63%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.54 - 0.62 (m, 2 H), 0.86 - 0.96 (m, 2 H), 1.53 - 1.63 (m, 1 H), 1.87 - 1.99 (m, 2 H), 2.78 - 2.90 (m, 2 H), 3.49 - 3.63 (m, 2 H), 6.98 (td, ./=8.5, 1.8 Hz, 1 H), 7.30 (d, 7=9.2 Hz, 1 H), 7.43 (dd, /=8.2, 6.9 Hz, 1 H), 7.64 (dt, 7=11.8, 2.2, 2.1 Hz, 1 H), 7.71 (s, 1 H), 8.02 (br., 3 H), 8.90 (br. s, 1 H), 10.83 (br. s, 1 H); m/z (ES+APCI)+: 302 [M+H]+.
Intermediate 4 Nl-(2-Chloro-5-cyclopropyl-pyrimidin-4-yl)-propane-l,3-diamine dihydrochloride
Figure imgf000064_0002
A suspension of
Intermediate 2 (2 g, 6.13 mmol) in 4M HCl in dioxane (15 ml, 60 mmol) was stirred vigorously for 1 hour at room temperature The reaction mixture was evaporated to dryness, azeotroped with toluene (25 ml) followed by DCM (40 ml) to give an off-white solid dihydrochloride salt 100% (1.61 g, 100%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.55 - 0.61 (2 H, m), 0.91 (2 H, m), 1.58 (1 H, m), 1.83 - 1.92 (2 H, m), 2.78 - 2.87 (2 H, m), 3.47 (2 H, m), 7.50 (1 H, br. s), 7.73 (1 H, s), 7.98 (1 H, t, ./=5.72 Hz), 8.08 (2 H, br. s); m/z (ES+APCI)+; 227 302 [M+H]+.
Intermediate 5
N-{3'[4-(3-Aminoψropylamino)-5-cyclopropyl-pyrimidin-2-ylammo]-phenyl}-acetamide dihydrochloride
Figure imgf000065_0001
Step 1
{3-[2-(3-Acetylaminoψhenylamino)-5-cyclopropyl-pyrirnidin-4-ylamino]-propyl}-carbamic acid tert-butyl ester
Figure imgf000065_0002
Intermediate 2 (200 mg, 0.61 mmol), N-(3-aminophenyl)-acetamide (184 mg, 1.22 mmol) and glacial acetic acid (7 μl, 0.12 mmol) were dissolved in n-butanol (3 ml). The reaction mixture was irradiated at 140 0C for 15 minutes in a Biotage 1-60 microwave reactor. The reaction was evaporated to dryness, dissolved in DCM (10 ml), partitioned with saturated sodium bicarbonate solution (10 ml) and separated. The aqueous was extracted with DCM (20 ml) and the combined organic layers were dried (MgSO4) and evaporated to dryness. The crude product was subjected to repeated flash chromatography on a Biotage SP4 (DCM/methanol gradient) to give an off-white solid (0.167 g). 1H ΝMR data are consistent with the desired product, but indicate the presence of approximately 20% of unreacted N-(3-Aminophenyl)-acetamide starting material impurity, m/z (ES+APCI)+: 441 [M+H]+. This product was used in the next step without further purification.
Step 2. N-{3-[4-(3-Amino-propylamino)-5-cyclopropyl-pyrimidin-2-ylamino]-phenyl}-acetamide dihydrochloride
A suspension of {3-[2-(3-Acetylamino-phenylamino)-5-cyclopropyl-pyrimidin-4-ylamino]- propyl}-carbamic acid
Figure imgf000066_0001
ester prepared in step 1 (167 mg, 0.38 mmol) in 4M hydrogen chloride in dioxane (1 ml) was stirred vigorously for 2 h at room temperature. The reaction was evaporated to dryness, triturated with DCM, filtered and dried to give the title compound as an off- white solid (169 mg, 74% over 2 steps). 1H ΝMR (400 MHz, DMSO-c/6) δ ppm 0.53 - 0.61 (m, 2 H), 0.84 - 0.96 (m, 2 H), 1.53 - 1.62 (m, 1 H), 1.85 - 1.95 (m, 2 H), 2.07 (s, 3 H), 2.78 - 2.87 (m, 2 H), 3.60 - 3.67 (m, 2 H), 7.18 - 7.23 (m, 2 H), 7.27 - 7.33 (m, 1 H), 7.67 (s, 1 H), 7.98 (br. s, 2 H), 8.12 (s, 1 H), 8.76 - 8.82 (m, 1 H), 10.16 (s, 1 H), 10.52 (s, 1 H); m/z (ES+APCI)+: 341 [M+H]+.
Intermediate 6
Thiophene-2-carboxylic
Figure imgf000066_0002
[3-(2<Moro-5-cyclopropyl-pyrimidin-4-ylamino)-propyl]-amide
Figure imgf000067_0001
To an ice cooled solution of Intermediate 4 (2.45g, 9.3 mmol) and triethylamine (2.8 ml, 20.45 mmol) in DCM was added thiophene-2-carbonyl chloride (1.02 ml, 9.5 mmol) dropwise. The reaction was stirred at room temperature for 2.5 h and then quenched with water (50 ml), and the aqueous layer was extracted with DCM (2 x 50 ml). The combined organic layers were washed with brine (50 ml), dried (MgSO4) and evaporated to dryness. The crude product was subjected to flash chromatography on the Biotage SP4 (gradient elution from 0 - 6% methanol in DCM) to give a foam, which was triturated with petroleum ether to give a white solid (1.56 g, 50%). 1H NMR (400 MHz, OMSO-d6) δ ppm 0.52 - 0.59 (m, 2 H), 0.84 - 0.93 (m, 2 H), 1.47 - 1.56 (m, 1 H), 1.74 - 1.84 (m, 2 H), 3.26 - 3.33 (m, 2 H), 3.39 - 3.46 (m, 2 H), 7.14 (dd, /=4.8, 3.9 Hz, 1 H), 7.45 - 7.51 (m, 1 H), 7.70 (s, 1 H), 7.71 - 7.76 (m, 2 H), 8.52 - 8.57 (m, 1 H); m/z (ES+APCI)+: 337 [M+H]+.
Intermediate 7 l-[3-(2-Chloro-5-cyclopropyl-pyriτnidin-4-ylamino)-propyl]-pyrrolidin-2-one
Figure imgf000067_0002
Intermediate 1 (200 mg, 1.06 mmol), N-(3-aminopropyl)-2-pyrrolidinone (148 μl, 1.06 mmol) and NN-diisopropylethylamine (564 μl, 3.17 mmol) were combined in isopropyl alcohol (1 ml) at 0 °C and then stirred at this temperature for 1 hour. After this time, the reaction mixture was warmed up to room temperature and stirring continued overnight. The reaction mixture was partitioned between water and ethyl acetate, the aqueous phase was extracted with ethyl acetate (x2), the combined organic extracts washed with water, followed by brine, dried over MgSO4 and solvents evaporated to dryness. The crude product was purified by flash chromatography on the Biotage
SP4 (gradient elution from 0 - 10% methanol in ethyl acetate) to give a brown oil (197 mg, 63%). 1H NMR (400 MHz, DMSO-Cf6) δ ppm 0.50 - 0.56 (m, 2 H), 0.80 - 0.88 (m, 2 H), 1.43 - 1.51 (m, 1 H), 1.60 - 1.72 (m, 2 H), 1.85 - 1.95 (m, 2 H), 2.14 - 2.23 (m, 2 H), 3.20 (t, J=6.9 Hz, 2 H), 3.33 (m, 4H), 7.44 (t, j=5.7 Hz, 1 H), 7.66 (s, 1 H); m/z (ES+APCI)+: 295 [M+H]+.
Intermediate 8
Cyclobutanecarboxylic acid [3-(2-chloro-5-cyclopropyl-pyrimidin-4-ylamino)-propyl]-amide
Figure imgf000068_0001
Prepared analogously to Intermediate 6 from Intermediate 4 and cyclobutane carbonyl chloride to give desired product as a colourless oil (0.83 g, 35%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.50 - 0.61 (m, 2 H), 0.82 - 0.94 (m, 2 H), 1.45 - 1.57 (m, 1 H), 1.60 - 1.68 (m, 2 H), 1.68 - 1.78 (m, 1 H), 1.80 - 1.93 (m, 1 H), 1.94 - 2.04 (m, 2 H), 2.05 - 2.17 (m, 2 H), 2.92 - 3.03 (m, 1 H), 3.05 - 3.12 (m, 2 H), 3.29 - 3.40 (m, 2 H), 7.44 - 7.50 (m, 1 H), 7.66 - 7.74 (m, 2 H). Rf (5% MeOH in DCM) = 0.4.
Alternatively intermediate 8 can be prepared according to the following method:
A solution of Intermediate 11 below (5.16 g, 19.04 mmol) in IPA (20 ml) was added to a stirred solution of intermediate 1 (3.6 g, 19.04 mmol) and DIPEA (13.2 ml, 72.2 mmol) in IPA (80 ml) with ice-cooling. The mixture was allowed to warm to rt and then heated at 55°C overnight. The mixture was allowed to cool to rt and concentrated. The residue was dispersed into ethyl acetate and water. The organic phase was dried and concentrated to give an oil. The crude oil was purified by flash column chromatography on silica gel (200 g) eluting with 30:1 DCM:MeOH to afford a white solid (4.3 g, 73 %).
Intermediate 9 Cyclopentanecarboxylic acid [3-(2-chloro-5-cyclopropyl-pyriτnidin-4-ylamino)-propyl] -amide
Figure imgf000069_0001
To a solution of Intermediate 4 (0.2g, 0.76 mmol) in DMF (2 ml) was added cyclopentane carboxylic acid (0.124 ml, 1.14 mmol), HATU (0.46 g, 1.22 mmol) and N,N-diisopropylethylamine
(0.79 ml, 4.56 mmol). The reaction mixture was stirred at room temperature for 18 hours. The mixture was evaporated and filtered through a 1 g Isolute-ΝH2 cartridge, eluting with 9: 1 DCM : methanol. Purification by flash chromatography on the Biotage SP4 (gradient elution from 0 to 5% methanol in DCM) gave the desired product as a colourless oil (45 mg, 18%). 1H NMR (400 MHz,
DMSCW6) δ ppm 0.53 - 0.58 (m, 2 H), 0.86 - 0.92 (m, 2 H), 1.46 - 1.54 (m, 3 H), 1.54 - 1.76 (m, 9
H), 3.06 - 3.13 (m, 2 H), 3.29 - 3.40 (m, 2 H), 7.44 - 7.49 (m, 1 H), 7.69 (s, 1 H), 7.79 - 7.84 (m, 1 H); m/z (ES+APCI)+: 323 [M+H]+.
Intermediate 10
Pyrrolidine-! -carboxylic acid (3-amino-phenyl)-amide
Figure imgf000069_0002
Stepl
N-(3-nitrophenyl)pyrrolidine-l-carboxamide
To a stirred solution of 3-nitrophenylisocyanate (10 g, 60.9 mmol) in dry THF (100 ml) at 0 0C, was added pyrrolidine (6.04 ml, 73.1 mmol), and the resulting solution was heated to reflux overnight. The solvent was removed under reduced pressure and the resulting yellow solid was triturated with ethyl acetate, filtered, washed with 1:1 ethyl acetate - petroleum ether and dried under vacuum. The product was isolated as a pale yellow solid (12.8 g, 89%). 1H NMR (400 MHz, DMSCW6) δ ppm 1.81-1.91 (m, 4 H), 3.35-3.43 (m, 4 H), 7.50 (t, 7=8.28 Hz, 1 H), 7.76 (ddd, 7=8.28, 2.76, 0.92 Hz, 1 H), 7.96 (ddd, 7=8.28, 1.8, 0.92 Hz, 1 H), 8.55 (t, 7=1.8 Hz, 1 H), 8.64 (br s, 1 H); m/z (ES+ APCI)" 235 [M-H]. Step 2
Pyrrolidine- 1-carboxylic acid (3-amino-phenyl)-amide
To a solution of N-(3-nitrophenyl)pyrrolidine-l-carboxamide prepared in step 1 (10 g, 42.6 mmol) in ethyl acetate (150 ml) and ethanol (10 ml) was added 10% Pd on activated carbon (1 g). The reaction was stirred under an atmosphere of hydrogen at room temperature for 18 h. More 10% Pd on activated carbon (0.4 g) was added and the reaction was complete after stirring for a further 4 hours. The mixture was filtered through Celite™, washing with ethanol, and then evaporated to give a pink foam, which was triturated with petroleum ether - EtOAc (10:1). The resultant solid was filtered and dried under vacuum to give the desired product as a pale pink solid (4.68 g, 54%). 1H NMR (400 MHz, DMS(W6) δ ppm 1.78 - 1.88 (m, 4 H), 3.25 - 3.39 (m, 4 H), 4.88 (br. s, 2 H), 6.14 (ddd, 7=7.0, 1.1, 1.0 Hz, 1 H), 6.61 (ddd, .7=7.0, 1.1, 1.0 Hz, 1 H), 6.76 - 6.89 (m, 2 H), 7.75 (br. s, 1 H). Rf (6% methanol: 94% DCM) = 0.35.
Intermediate 11
Cyclobutanecarboxylic acid (3-amino-propyl) -amide trifluoroacetate salt
Figure imgf000070_0001
Step l
[i-iCyclobutanecarbonyl-amino) -propyl] -carbamic acid tert-butyl ester:
A solution of cyclobutanecarbonyl chloride (6.7 ml, 58.6 mmol) in DCM (20 ml) was added slowly to a stirred solution of (3-amino-propyl)-carbamic acid tert-butyl ester (10.0 g, 57.5 mmol) and TEA (12.0 ml, 86.2 mmol) in DCM (180 ml) with ice cooling. The RM was then stirred at rt for 2 hours. After this time the mixture was washed with 10% aqueous citric acid and water. The organic phase was separated, dried and concentrated to provide a white solid (14.3 g, 97 %). 1H NMR (400 MHz, DMSCW6) δ ppm 1.37 (9 H, m), 1.40 - 1.55 (2 H, m), 1.62 - 2.30 (6 H, m), 2.75 - 3.12 (5 H, m), 6.78 (1 H, t, /=5.50 Hz), 7.61 (1 H, t, /=5.50 Hz) Step 2
Cyclobutanecarboxylic acid (3-amino-propyl) -amide trifluoroacetate salt
[3-(Cyclobutanecarbonyl-amino)-propy]]-carbamic acid tert-buty\ ester (1 1.3 g, 44.0 mmol) in TFA (20 ml) and DCM (60 ml) was stirred at it for 5 hours. The mixture was then concentrated to dryness. Toluene was added to the residue and the mixture concentrated to dryness again to provide a viscous oil (22.2 g). 1H NMR (400 MHz, DMSCW6) δ ppm 1.59 - 2.18 (8 H, m), 2.60 - 2.85 (2 H, m), 2.85 - 3.14 (3 H, m), 7.60 - 7.91 (3 H, m); m/z (ES+APCI)+ : 157 [M+H]+.
Intermediate 12
N,N,N'-Trimethyl-N'-(5-nitro-pyridin-2-yl)-ethane-l,2-diamine
Figure imgf000071_0001
N,N,N'-Trimethylethylenediamine (798 μl, 6.29 mmol) was added to 2-chloro-5-nitropyridine (1.0 g, 6.29 mmol) and DIPEA (1.09 ml, 6.29 mmol) in MeCN (20 ml). The mixture was stirred at rt for
4 hours and then concentrated to dryness. The residue was dispersed into DCM and saturated aqueous sodium carbonate solution. The organic phase was dried and concentrated to give an orange coloured oily solid (1.3 g, 92 %). 1H NMR (400 MHz, CHLOROFORM-c?) δ ppm 2.30 (6 H, s), 2.49 - 2.57 (2 H, m), 3.19 (3 H, s), 3.79 (2 H, br. s.), 6.46 (1 H, d, 7=9.62 Hz), 8.12 - 8.22 (1 H, m), 8.92 - 9.08 (1 H, m).
Intermediate 13
N -(2-Dimethylamino-ethyl)-N -methyl-pyridine-2,5-diamine
Figure imgf000071_0002
A mixture of Intermediate 12 (1.3 g, 5.8 mmol) and 10 % Pd/C (130 mg) in EtOH (40 ml) were stirred at it under an atmosphere of hydrogen for 4 hours. The mixture was then filtered through Celite and the filtrate concentrated to dryness to give a deep purple coloured oil. The crude oil was purified by flash column chromatography on silica gel (60 g) eluting with 10: 1 DCM:2M NH3 in MeOH to provide a deep purple coloured oil (470 mg, 42%). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.14 (6 H, s), 2.27 - 2.34 (2 H, m), 2.84 (3 H, s), 3.41 - 3.49 (2 H, m), 4.35 (2 H, br. s.), 6.39 (1 H, d, /=8.70 Hz), 6.88 (1 H, dd, /=8.93, 2.98 Hz), 7.54 (1 H, d, J=2.29 Hz); m/z (ES+APCI)+ : 195 [M+H]+.
Intermediate 14
2-[Methyl-(5-nitro-pyridin-2-yl)-amino]-ethanol
Figure imgf000072_0001
Prepared analogously to Intermediate 12 from 2-chloro-5-nitropyridine and 2-methylamino-ethanol to give an orange coloured oil (1.32 g, 106%). 1H NMR (400 MHz, DMSO-J6) δ ppm 3.19 (3 H, s), 3.51 - 3.83 (4 H, m), 4.83 (1 H, br. s.), 6.76 (1 H, br. s.), 8.19 (1 H, dd, /=9.62, 2.29 Hz), 8.95 (1 H, d, /=2.75 Hz)
Intermediate 15 2-[(5-Amino-pyridin-2-yl) -methyl-aminoj-ethanol
Figure imgf000072_0002
Prepared analogously to Intermediate 13 from Intermediate 14 to give a deep purple coloured oil (912 mg, 83 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.89 (3 H, s), 3.37 - 3.54 (4 H, m), 4.36 (2 H, s), 4.66 (1 H, t, /=5.04 Hz), 6.42 (1 H, d, /=8.70 Hz), 6.89 (1 H, dd, /=8.93, 2.98 Hz), 7.53 (1 H, d, /=2.75 Hz); m/z (ES+APCI)+ : 168 [M+H]+. Intermediate 16
Methyl-(2-morpholin-4-yl-ethyl)-(5-nitro-pyridin-2-yl)-amine
Figure imgf000073_0001
Prepared analogously to Intermediate 12 from 2-chloro-5-nitropyridine and methyl-(2-morpholin- 4-yl-ethyl)-amine to give an orange oil (790 mg, 94 %). 1H NMR (400 MHz, CHLOROFORM-./) δ ppm 2.41 - 2.65 (6 H, m), 3.21 (3 H, s), 3.70 (4 H, m), 3.83 (2 H, br. s.), 6.47 (1 H, d, 7=9.16 Hz), 8.21 (1 H, dd, 7=9.39, 2.98 Hz), 9.05 (1 H, d, 7=2.75 Hz)
Intermediate 17
N -Methyl-N -(2-morpholin-4-yl-ethyl)-pyridine-2,5-diamine
Figure imgf000073_0002
Prepared analogously to Intermediate 13 using Intermediate 16 to give a deep purple coloured oil
(352 mg, 50%). 1H NMR (400 MHz, CHLOROFORM-rf) δ ppm 2.41 - 2.65 (6 H, m), 2.99 (3 H, s), 3.55 - 3.75 (6 H, m), 6.42 (1 H, d, /=9.62 Hz), 6.99 (1 H, dd, J=8.70, 2.75 Hz), 7.76 (1 H, d, 7=2.29 Hz); m/z (ES+APCI)+ : 237 [M+H]+. Intermediate 18
5-Amino-pyridine-2-carboyylic acid ethyl ester
Thionyl chloride (5.2 ml, 71.3 mmol) was added dropwise to 5-amino-pyridine-2-carboxylic acid
(4.0 g, 29.0 mmol) in EtOH (30 ml) whilst maintaining the temperature of the mixture below 100C. During the addition the mixture became very thick and more EtOH (20 ml) was added. The mixture was then stirred and heated at 95°C overnight. Next day the temperature was increased to 1 100C and the mixture was heated at this temperature overnight. On cooling to rt the mixture was concentrated to give a solid. Saturated aqueous sodium hydrogen carbonate solution was added followed by saturated aqueous sodium carbonate solution. Ethyl acetate and water were then added. The organic phase was washed with brine, dried and concentrated to give an off white solid (4.0 g, 83 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.27 (3 H, t, /=7.10 Hz), 4.22 (2 H, q, 7=6.87 Hz), 6.19 (2 H, s), 6.91 (1 H, dd, 7=8.70, 2.75 Hz), 7.74 (1 H, d, J=8.70 Hz), 7.97 (1 H, d, J=2.29 Hz).
Intermediate 19
(5 -Amino-pyridin-2-yl) -methanol
Figure imgf000074_0001
Lithium aluminium hydride, 2 M in THF (29.8 ml, 59.6 mmol) was added slowly to a solution of Intermediate 18 (3.3 g, 19.9 mmol) in THF (120 ml) with ice-cooling. The mixture was stirred at rt for 4 hours. Water (2.3 ml) was then added dropwise whilst maintaining the temperature of the mixture below 1O0C. An aqueous solution of sodium hydroxide, 15 % w/v (2.3 ml) was subsequently added followed by an additional quantity of water (6.9 ml). The resulting mixture was filtered and the filtrate concentrated to provide an orange coloured solid (2.59 g, 105%). 1H NMR (400 MHz, DMSO-J6) δ ppm 4.35 (2 H, d, 7=5.50 Hz), 5.04 (1 H, m), 5.16 (2 H, s), 6.90 (1 H, dd, 7=8.24, 2.75 Hz), 7.07 (1 H, d, 7=8.24 Hz), 7.82 (1 H, d, 7=2.75 Hz).
Intermediate 20 6-(tert-Butyl-dimethyl-silanyloxymethyl)-pyridin-3-ylamine
Figure imgf000074_0002
A mixture of Intermediate 19 (1.43g, 11.5 mmol), TBDMSCl (2.09 g, 13.8 mmol) and imidazole (1.88 g, 27.7 mmol) in DMF (30 ml) were stirred at rt overnight. The mixture was diluted with EtOAc, washed with water (x 4) and brine (x 1). The organic phase was dried and concentrated. The crude residue was purified by flash column chromatography on silica gel (100 g) eluting with 2:1 ethyl acetate:petrol to give an off-white solid (1.72 g, 63 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.04 (6 H, s), 0.88 (9 H, s), 4.55 (2 H, s), 5.20 (2 H, s), 6.91 (1 H, dd, 7=8.24, 2.75 Hz), 7.06 (1 H, d, 7=8.24 Hz), 7.83 (1 H, d, 7=2.75 Hz). Intermediate 21
Cyclobutanecarboxylic acid (3-{2-[6-(tert-butyl-dimethyl-silanyloxymethyl)-pyridin-3-ylamino]-5- cyclopropyl-pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000075_0001
A mixture of Intermediate 20 (1.72 g, 7.23 mmol), Intermediate 8 (1.11 g, 3.61 mmol) and AcOH (206 μl, 3.61 mmol) in H-BuOH (20 ml) were stirred and heated at 115°C overnight. On cooling to rt the mixture was concentrated to dryness. The residue was taken up in EtOAc and washed with saturated aqueous sodium hydrogen carbonate solution and brine. The organic phase was dried and concentrated. The crude residue was purified by flash column chromatography on silica gel (150 g) eluting with a MeOH-EtOAc gradient (2 % to 5 % MeOH) to give an off-white solid (590 mg, 32 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.08 (6 H, s), 0.45 - 0.50 (2 H, m), 0.81 - 0.86 (2 H, m), 0.91 (9H, s), 1.44 - 1.51 (1 H, m), 1.66 - 1.78 (3 H, m), 1.82 - 1.90 (1 H, m), 1.94 - 2.03 (2 H, m), 2.06 - 2.17 (2 H, m), 2.93 - 3.00 (1 H, m), 3.10 - 3.17 (2 H, m), 3.39 - 3.46 (2 H, m), 4.66 (2 H, s), 6.90 (1 H, t, 7=5.95 Hz), 7.29 (1 H, d, J=8.24 Hz), 7.62 (1 H, s), 7.69 (1 H, t, /=5.72 Hz), 8.21 (I H, dd, ./=8.47, 2.52 Hz), 8.81 (1 H, d, J=2.29 Hz), 9.08 (1 H, s); m/z (ES+APCI)+ : 511 [M+H]+.
Intermediate 22
Cyclobutanecarboxylic acid {3-[2-(6-chloromethyl-pyridin-3-ylamino)-5-cyclopropyl-pyrimidin-4- ylamino]-propyl}-amide x.HCl
Figure imgf000075_0002
Thionyl chloride (194 μl, 2.66 mmol) was added dropwise to a suspension of Example 103 (211 mg, 0.533 mmol) in DCM (20 ml) with ice-cooling. The mixture was allowed to warm to rt and stirred at this temperature overnight. The mixture was then concentrated to dryness to give an off- white solid as the x.HCl salt (274 mg). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.55 - 0.60 (2 H, m), 0.88 - 0.94 (2 H, m), 1.52 - 1.60 (1 H, m), 1.65 - 1.75 (3 H, m), 1.79 - 1.99 (3 H, m), 2.02 - 2.12 (2
H, m), 2.92 - 2.98 (1 H, m), 3.06 - 3.17 (2 H, m), 3.42 - 3.49 (2 H, m), 4.79 (2 H, s), 7.61 (1 H, d, 7=8.70 Hz), 7.66 (1 H, s), 7.80 (1 H, t, 7=5.72 Hz), 8.07 (1 H, dd, 7=8.47, 2.52 Hz), 8.73 - 8.79 (2 H, m), 10.79 (1 H, s); m/z (ES+APCI)+ : 415/417 [M+H]+.
Intermediate 23
[3-(2-Chloro-5-cyclopropyl-pyriτnidin-4-yloxy)-propyl]-carbamic acid tert-butyl ester
Figure imgf000076_0001
3-(Boc-amino)-l-propanol (278 mg, 1.59 mmol) dissolved in dry THF (1 ml) was added dropwise to a stirred solution of sodium hydride (60% dispersion in mineral oil, 38 mg, 1.59 mmol) in dry
THF (1 ml) at 0 0C under nitrogen. The solution was stirred at 0 0C for 30 minutes, then
Intermediate 1 (300 mg, 1.59 mmol), in dry THF (1 ml) was added dropwise and stirring continued for further 20 minutes. The reaction mixture was then warmed up to room temperature and stirred at this temperature overnight. The reaction mixture was partitioned between water and DCM, the organic extract was washed with saturated aqueous NaHCO3, followed by brine, dried (MgSO4) and solvents removed. Purification by column chromatography using a Biotage SP4 (petroleum ether/ethyl acetate gradient) gave the product as a clear oil (292 mg, 56%). 1H NMR (400 MHz,
CHLOROFORM-^ δ ppm 0.63 - 0.79 (m, 2 H), 0.91 - 1.07 (m, 2 H), 1.44 (s, 9 H), 1.82 - 1.90 (m,
1 H), 1.93 - 2.06 (m, 2 H), 3.21 - 3.37 (m, 2 H), 4.51 (t, 7=6.0 Hz, 2 H), 7.94 (s, 1 H). Rf (8:2, petroleum ether/ethyl acetate) = 0.24.
Intermediate 24
{3-[5-Cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-yloxy]-propyl}-carbamic acid tert-butyl ester
Figure imgf000077_0001
Intermediate 23 (100 mg, 0.31 mmol), 3-fluoroaniline (59 μl, 0.61 mmol) and glacial acetic acid
(3.5 μl, 0.06 mmol) were dissolved in π-butanol (1 ml) and irradiated at 150 0C for 15 minutes in a
Biotage 1-60 microwave reactor. The reaction mixture was concentrated and the crude material purified by column chromatography on Biotage SP4 (petroleum ether/ethyl acetate gradient) to give the product as a white solid (25 mg, 20%). 1H NMR (400 MHz, DMSOd6) δ ppm 0.64 - 0.69
(m, 2 H), 0.79 - 0.84 (m, 2 H), 1.35 (s, 9 H), 1.72 - 1.83 (m, 1 H), 1.83 - 1.93 (m, 2 H), 3.04 - 3.17
(m, 2 H), 4.38 (t, J=6.0 Hz, 2 H), 6.69 (td, /=8.2, 2.3 Hz, 1 H), 6.91 (t, /=5.5 Hz, 1 H), 7.22 - 7.30
(m, 1 H), 7.46 (d, /=8.2 Hz, 1 H), 7.77 (d, /=12.8 Hz, 1 H), 7.94 (s, 1 H), 9.60 (s, 1 H); m/z (ES+APCI)+: 402 [M]+.
Intermediate 25 ft-β-Amino-propoxyj-S-cyclopropyl-pyrimidin^-ylJ-β-fluoro-phenylj-amine hydrochloride salt
Figure imgf000077_0002
4M HCl in dioxane (1 ml) was added to Intermediate 24 (20 mg, 0.05 mmol) and the reaction mixture stirred at room temperature for 4 hours. The solvents were removed to give the product as a white solid (17 mg, 100%). 1H NMR (400 MHz, DMSOd6) δ ppm 0.65 - 0.71 (m, 2 H), 0.75 - 0.89 (m, 2 H), 1.74 - 1.82 (m, 1 H), 2.03 - 2.14 (m, 2 H), 2.91 - 3.02 (m, 2 H), 4.48 (t, /=6.0 Hz, 2 H), 6.77 (td, J=8.4, 2.1 Hz, 1 H), 7.26 - 7.34 (m, 1 H), 7.44 (d, /=8.2 Hz, 1 H), 7.73 (dd, 7=12.4,
2.3 Hz, 1 H), 7.99 (s, 2 H), 8.01 (br. s, 2 H), 9.98 (br. s, 1 H); m/z (ES+APCI)+: 303 [M+H]+.
Intermediate 26
Cyclobutanecarboxylic acid (3-methylamino-propyl)-amide hydrochloride salt
Figure imgf000078_0001
Step l β-fCyclobutanecarbonyl-amino) -propyl] '-methyl-carbamic acid tert-butyl ester To a stirred solution of N-(3-Aminopropyl)-N-methylcarbamic acid tert-butylester (600 mg, 3.19 mmol) under nitrogen at 0 0C was added N,N-diisopropylethylamine (610 μl, 3.51 mmol), followed by a dropwise addition of cyclobutanecarbonyl chloride (366 μl, 3.19 mmol). The reaction mixture was stirred at 0 0C for 1 hour, then allowed to warm up to room temperature and stirring continued overnight. The solution was diluted with DCM and washed successively with saturated aqueous NaHCO3, IM HCl (aq), water and brine, and the organic phase was dried (MgSO4) and concentrated. Purification by column chromatography on Biotage SP4 (ethyl acetate/methanol gradient) gave a colourless oil (281 mg, 49%) which was used in Step 2 without further purification.
Step 2 Cyclobutanecarboxylic acid (3-methylamino-propyl)-amide hydrochloride salt
4M HCl in dioxane (2 ml) was added to [3-(Cyclobutanecarbonyl-amino)-propyl]-methyl- carbamic acid tert-butyl ester (206 mg, 0.76 mmol) and the reaction mixture stirred for 3 hours at room temperature. The solvents were removed to give the title compound as a white solid (120 mg, 76%). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.65 - 1.79 (m, 3 H), 1.83 - 1.93 (m, 1 H), 1.95 - 2.05 (m, 2 H), 2.05 - 2.17 (m, 2 H), 2.73 - 2.89 (m, 2 H), 2.98 (quin, J=8.36, 8.24, 8.01 Hz, 1 H), 3.05 - 3.12 (m, 2 H), 7.89 (t, 7=5.72 Hz, 1 H), 8.75 (br. s, 2 H); m/z (ES+APCI)+: 171 [M+H]+. Intermediate 27
Cyclobutanecarboxylic acid {3-[(2-chloro-5-cyclopropyl-pyrimidin-4-yl)-methyl-amino]-propyl}- amide
Figure imgf000079_0001
Intermediate 1 (103 mg, 0.54 mmol),
Intermediate 26 (1 12 mg, 0.54 mmol) and N,N-diisopropylethylamine (289 μl, 1.63 mmol) were combined in isopropanol at 0 0C, stirred at this temperature for 45 minutes, warmed up to room temperature, stirred for 10 minutes and then heated to 55 0C overOnight. The reaction mixture was allowed to cool to room temperature and partitioned between water and ethyl acetate, extracted twice with ethyl acetate and the combined organic extracts washed with brine, dried (MgSO4) and solvents removed. Purification by column chromatography on Biotage SP4 (petroleum ether/ethyl acetate gradient) gave the product as a clear oil (120 mg, 69%). 1H ΝMR (400 MHz, DMSO-^6) δ ppm 0.65 - 0.71 (m, 2 H), 0.78 - 0.96 (m, 2 H), 1.63 - 1.77 (m, 3 H), 1.81 - 2.02 (m, 4 H), 2.03 - 2.14 (m, 2 H), 2.94 (quin, J=8.24 Hz, 1 H), 3.00 - 3.08 (m, 2 H), 3.12 - 3.28 (m, 3 H), 3.46 - 3.72 (m, 2 H), 7.66 (t, /=5.50 Hz, 1 H), 7.88 (s, 1 H); m/z (ES+APCI)+: 323 [M+H]+.
Intermediate 28
[3-(2-Chloro-5-cyclopropyl-pyrimidin-4-ylamino)-propyl]-methyl-carbamic acid tert-butyl ester
Figure imgf000079_0002
Intermediate 1 (600 mg, 3.2 mmol) was dissolved in isopropyl alcohol and the solution was cooled to 0 0C, then N-(3-aminopropyl) carbamic acid tert-buty\ ester (450 mg, 3.12 mmol) was added, followed by N,N-diisopropylethylamine (1.69 ml, 9.52 mmol). The solution was stirred for 1 h, warmed to room temperature, stirred for 20 min. and, then warmed to 55 0C and stirred overnight. The reaction mixture was partitioned between ethyl acetate and water, extracted twice with ethyl acetate and the combined organic extracts washed with brine, dried (MgSO4) and solvents evaporated to dryness. The crude product was purified by flash chromatography using a Biotage SP4 (ethyl acetate / petroleum ether gradient) to give the product as a clear oil (602 mg, 56%). 1H ΝMR (400 MHz, DMSO-^6, 40 0C) δ ppm 0.57 - 0.62 (m, 2 H), 0.87 - 0.96 (m, 2 H), 1.41 (br. s, 9 H), 1.48 - 1.60 (m, 1 H), 1.81 (br. s, 2 H), 2.82 (s, 3 H), 3.14 - 3.33 (m, 2 H), 3.40 (q, 7=6.9 Hz, 2 H), 7.38 (br. s, 1 H), 7.73 (s, 1 H); m/z (ES+APCI)+: 241 [M - CO2'Bu+H], 341 [M+H]+.
Intermediate 29
5-Cyclopropyl-N2-(3-fluoro-phenyl)-N4-(3-methylamino-propyl)-pyrimidine-2,4-diamine hydrochloride salt
Figure imgf000080_0001
Intermediate 28 (300 mg, 0.9 mmol) was dissolved in 14:1 acetonitrile : water (435 ml) and to it added 3-fluoroaniline (110 μl, 1.2 mmol), followed by 4 M HCl in dioxane (320 μl), and the reaction mixture was heated to 50 0C overnight. The solvents were evaporated and the crude product was purified by flash chromatography using a Biotage SP4 (DCM / 3% ammonia in methanol gradient) to give the product as a yellow solid (236 mg, 85%). 1H ΝMR (400 MHz, DMSO-40 δ ppm 0.60 - 0.65 (m, 2 H), 0.89 - 1.02 (m, 2 H), 1.60 - 1.67 (m, 1 H), 1.96 - 2.08 (m, 2 H), 2.50 (s, 3H, obscured by DMSO peak), 2.81 - 3.00 (m, 2 H), 3.51 - 3.72 (m, 2 H), 6.52 - 6.65 (m, 1 H), 7.03 (td, J=8.2, 2.3 Hz, 1 H), 7.29 - 7.38 (m, 1 H), 7.42 - 7.55 (m, 1 H), 7.55 - 7.71 (m, 1
H), 7.76 (s, 1 H), 8.92 - 9.00 (m, 1 H), 11.00 (s, 1 H); m/z (ES+APCI)+: 316 [M+H]+.
Intermediate 30 l-(3-Amino-propyl)-piperidin-2-one.hydrochloride salt.
Figure imgf000081_0001
Step l
[3-(2-Oxo-piperidin-l-yl)-propyl]-carbamic acid tert-butyl ester
Figure imgf000081_0002
Sodium hydride (60% dispersion in mineral oil; 425 mg, 10.6 mmol) was added to anhydrous THF
(10 ml), under nitrogen and the mixture cooled down to 0 0C. A solution of valerolactam (500 mg, 5.1 mmol) in anhydrous THF (10 ml) was added dropwise and the reaction mixture stirred at this temperature for 30 min. After this time, a solution of tert-butyl (3-bromopropyl)carbamate (1.44 g, 6.1 mmol) in anhydrous THF (10 ml) was added dropwise, stirring at 0 0C continued for further 15 min., then the solution allowed to warm up to room temperature and stirred overnight. The solution was cooled to 0 0C, saturated sodium hydrogen carbonate added and the aqueous phase extracted twice with DCM. The combined organic extracts were washed with brine, dried (MgSO4) and solvents removed to give the product as a clear oil (960 mg, 74%). The crude material was proceeded through to the next step without further purification. 1H NMR (400 MHz, DMSO-^6) δ ppm 1.42 (s, 9 H) 1.65 - 1.83 (m, 4 H) 2.12 - 2.26 (m, 2 H) 2.87 - 2.96 (ra, 2 H) 3.19 - 3.35 (m, 4 H) 4.14 - 4.20 (m, 2 H) 6.78 (t, 7=5.5 Hz, 1 H); m/z (ES+APCI)+: 157 [M - CO2'Bu+H].
Step 2 l-(3-Amino-propyl)-piperidin-2-one.hydrochloride salt.
4 M HCl in dioxane (6 ml) was added to [3-(2-Oxo-piperidin-l-yl)-propyl]-carbamic acid tert- butyl ester (910 mg, 3.6 mmol) and the reaction mixture was stirred at room temperature for 4 hours. The solvents were evaporated to give the title compound as an oily white solid (984 mg). 1H
NMR (400 MHz, DMSO-Uf6) δ ppm 1.76 - 1.92 (m, 4 H), 2.26 (t, /=6.2 Hz, 2 H), 2.69 - 2.80 (m, 2 H), 3.14 - 3.22 (m, 2 H), 3.27 (t, 7=5.7 Hz, 2 H), 3.36 (t, 7=6.9 Hz, 2 H), 8.03 (br. s, 3 H); m/z (ES+APCI)+ : 157 [M+H]+
Intermediate 31 l-[3-(2-Chloro-5-cyclopropyl-pyrimidin-4-ylamino)-propyl]-piperidin-2-one
Figure imgf000082_0001
Intermediate 1 (542 mg, 2.9 mmol),
Intermediate 30 (552 mg, 2.9 mmol) and N,N-diisopropylethylamine (1.53 ml, 8.6 mmol) were mixed together in isopropyl alcohol (10 ml) at 0 0C, stirred for 1 h, then warmed up to room temperature, stirred for 10 min., then warmed up to 55 0C overnight. The reaction mixture was partitioned between water and ethyl acetate, extracted twice with ethyl acetate, combined organic extracts washed with brine, dried (MgSO4) and solvents evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (ethyl acetate/methanol gradient) to give the product as a clear oil (401 mg, 45%). 1H ΝMR (400 MHz, DMSO-^6) δ ppm 0.54 - 0.63 (m, 2 H), 0.86 - 0.99 (m, 2 H), 1.50 - 1.59 (m, 1 H), 1.66 - 1.84 (m, 6 H), 2.26 (t, 7=6.2 Hz, 2 H), 3.30 (t, 7=6.0 Hz, 2 H), 3.35 - 3.41 (m, 4 H), 7.57 (t, 7=6.2 Hz, 1 H), 7.74 (s, 1 H); m/z (ES+APCI)+: 309 [M+H]+
Intermediate 32 l-(3-Amino-propyl)-3-methyl-lH-pyridin-2-one hydrochloride salt
Figure imgf000082_0002
[3-(3-Methyl-2-oxo-2H-pyridin-l-yl)-propyl]-carbamic acid tert-butyl ester
Figure imgf000083_0001
Sodium hydride (60% dispersion in mineral oil; 231 mg, 9.6 mmol) was added to anhydrous THF (10 ml), under nitrogen and the mixture cooled down to 0 °C. A solution of 3-methyl-2-pyridone (500 mg, 4.6 mmol) in anhydrous THF (10 ml) was added dropwise and the reaction mixture stirred at this temperature for 30 min. After this time, a solution of tert-butyl (3- bromopropyl)carbamate (1.31 g, 5.5 mmol) in anhydrous THF (10 ml) was added dropwise, and stirring was continued at 0 0C for a further 15 min., and then at room temperature overnight. The solution was cooled to 0 0C and then quenched with saturated sodium hydrogen carbonate(aq) and the aqueous phase extracted twice with DCM. The combined organic extracts were washed with brine and the solution filtered through a phase separation cartridge and solvents evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (ethyl acetate/methanol gradient) to give the product (165 mg, 14%). 1H NMR (400 MHz, CHLOROFORM-J) δ ppm 1.43 (s, 9 H), 1.84 - 1.93 (m, 2 H), 2.16 (s, 3 H), 3.04 - 3.12 (m, 2 H), 4.03 (t, 7=6.6 Hz, 2 H), 6.15 (t, /=6.9 Hz, 1 H), 7.22 (d, 7=6.4 Hz, 1 H), 7.18 (d, 7=6.9 Hz, 1 H); m/z (ES+APCI)+: 167 [M - CO2'Bu+H]+
Step 2
4 M HCl in dioxane (1 ml) was added to [3-(3-Methyl-2-oxo-2H-pyridin-l-yl)-propyl]-carbamic acid tert-butyl ester (160 mg, 0.6 mmol) and the reaction mixture stirred at room temperature for 3 hours. The solvents were evaporated to give the product as a brown solid (1 10 mg, 90%). 1H NMR
(400 MHz, DMSCW6) δ ppm 1.94 - 2.02 (m, 2 H), 2.04 (s, 3 H), 2.74 - 2.86 (m, 2 H), 4.03 (t,
7=6.9 Hz, 2 H), 6.23 (t, J=6.9 Hz, 1 H), 7.37 (d, 7=6.9 Hz, 1 H), 7.65 (d, 7=5.5 Hz, 1 H), 8.1 1 (br. s, 3 H); m/z (ES+APCI)+: 167 [M+H]+
Intermediate 33 l-[3z(2-Chloro-5-cyclopropyl-pyrimidin-4-ylaτnino)-propyl]-3-methyl-lH-pyridin-2-one
Figure imgf000084_0001
Intermediate 1 (103 mg, 0.5 mmol), Intermediate 32 (110 mg, 0.5 mmol) and N1N- diisopropylethylamine (290 μl, 1.63 mmol) were mixed together in isopropyl alcohol (1 ml) at 0 0C, stirred for 30 min., then warmed up to room temperature, stirred for 15 min., then warmed up to 55 °C overnight. The reaction mixture was partitioned between water and ethyl acetate, extracted twice with ethyl acetate and the combined organic extracts were washed with brine, dried (MgSO4) and solvents evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (petroleum ether/ethyl acetate) to give the product as a clear oil (120 mg, 64%). 1H NMR (400 MHz, DMSO-Cf6) δ ppm 0.55 - 0.67 (m, 2 H), 0.87 - 0.96 (m, 2 H), 1.52 - 1.59 (m, 1 H), 1.89 - 1.99 (m, 2 H), 2.03 (s, 3 H), 3.38 - 3.46 (m, 2 H), 3.94 - 4.03 (m, 2 H), 6.19 (t, J=6.9 Hz, 1 H), 7.33 (d, J=6.0 Hz, 1 H), 7.54 - 7.63 (m, 2 H), 7.74 (s, 1 H); m/z (ES+APCI)+: 319 [M+H]+
Intermediate 34 l-(3-Amino-propyl)-3-methyl-imidazolidin-2-one
Figure imgf000085_0001
Step l
[3-(3-Methyl-2-oxo-imidazolidin-l-yl)-propyl]-carbamic acid tert-butyl ester
Figure imgf000085_0002
Sodium hydride (60% dispersion in mineral oil; 252 mg, 10.5 mmol) was added to anhydrous THF (10 ml), under nitrogen and the mixture cooled down to 0 0C. A solution of l-methyl-2- imidazolidinone (500 mg, 5.0 mmol) in anhydrous THF (10 ml) was added dropwise and the reaction mixture stirred at this temperature for 30 min. After this time, a solution of tert-butyl (3- bromopropyl)carbamate (1.43 g, 6.0 mmol) in anhydrous THF (10 ml) was added dropwise at 0 0C and stirring at this temperature continued for further 15 min., before allowing to warm up to room temperature overnight. The solution was cooled to 0 0C and quenched with and water. Saturated sodium hydrogen carbonate(aq) was added and the aqueous phase was extracted twice with DCM. The combined organic extracts were washed with brine, dried (MgSO4) and solvents evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (ethyl acetate/methanol gradient) to give the product as a clear oil (279 mg, 22%). 1H NMR (400 MHz, CHLOROFORM-rf) δ ppm 1.43 (s, 9 H), 1.58 - 1.68 (m, 2 H), 2.78 (s, 3 H), 3.11 (t, 7=6.4 Hz, 2 H), 3.24 (t, 2 H), 3.29 (s, 4 H); m/z (ES+APCI)+: 158 [M - CO2'Bu+H]+
Step 2 l-(3-Amino-propyl)-3-methyl-imidazolidin-2-one
4 M HCl in dioxane (2 ml) was added to [3-(3-Methyl-2-oxo-imidazolidin-l-yl)-propyl]-carbamic acid tert-buty\ ester (269 mg, 1.1 mmol) and the reaction mixture stirred at room temperature for 1 h. The solvents were evaporated to give the product as a white solid (206 mg, 100%). 1H NMR (400 MHz, DMSCW6) δ ppm 1.61 - 1.88 (m, 2 H), 2.67 (s, 3 H), 2.71 - 2.81 (m, 2 H), 3.15 (t, ./=6.9 Hz, 2 H), 3.27 (s, 4 H), 7.93 (br. s, 3 H); m/z (ES+APCI)+: 158 [M+H]+ Intermediate 35 l-[3-(2-Chloro-5-cyclopropyl-pyrimidin-4-ylamino)-propyl]-3-methyl-imidazolidin-2-one
Figure imgf000086_0001
Intermediate 1 (195 mg, 1.0 mmol), Intermediate 34 (200 mg, 1.0 mmol) and NN- diisopropylethylamine (551 μl, 3.1 mmol) were mixed together in isopropyl alcohol (1 ml) at 0 0C, stirred for 30 min., then warmed up to room temperature and stirred for 15 min., then warmed up to 55 0C overnight. The reaction mixture was partitioned between water and ethyl acetate, extracted twice with ethyl acetate and the combined organic extracts were washed with brine, dried (MgSC^) and solvents evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (petroleum ether/ethyl acetate) to give the product as a white solid (159 mg, 50%). 1H ΝMR (400 MHz, DMSO--4) δ ppm 0.39 - 0.68 (m, 2 H), 0.88 - 0.97 (m, 2 H), 1.50 - 1.59 (m, 1 H), 1.69 - 1.80 (m, 2 H), 2.68 (s, 3 H), 3.17 (t, ./=6.9 Hz, 2 H), 3.24 - 3.35 (m, 4 H), 3.38 - 3.43 (m, 2 H), 7.52 (t, ^6.0 Hz, 1 H), 7.73 (s, 1 H); m/z (ES+APCI)+: 310 [M+H]+
Intermediate 36 l-(3-Amino-propyl)-3,3-dimethyl-pyrrolidin-2-one hydrochloride salt
Figure imgf000086_0002
Step 1
2-Oxo-pyrrolidine-l-carboxylic acid tert-butyl ester
O O Vf To a solution of 2-pyrrolidinone (3 g, 35.3 mmol) in DCM (80 ml) was added sequentially triethylamine (4.9 ml, 35.3 mmol), di- tert-buty\ dicarbonate (15.4 g, 70.6 mmol), and 4- dimethylaminopyridine (4.3 g, 35.3 mmol). The reaction mixture was stirred over 3 days, the solvents removed and the crude material purified by flash chromatography using a Biotage SP4 (ethyl acetate/methanol gradient) to give the product as a yellow oil (5.4 g, 83%). 1H NMR (400 MHz, CHLOROFORM-^) δ ppm 1.51 (s, 9 H), 1.93 - 2.04 (m, 2 H), 2.45 - 2.54 (m, 2 H), 3.68 - 3.77 (m, 2 H); Rf(1 :9 MeOH/EtOAc) = 0.6.
Step 2 3, 3-Dimethyl-2-oxo-pyrrolidine-l-carboxylic acid tert-butyl ester
O
2-Oxo-pyrrolidine-l-carboxylic acid tert-butyl ester (2 g, 1-0.8 mmol) was dissolved in anhydrous THF (100 ml), under nitrogen, cooled down to -78 0C and to it added LHMDS (1 M solution in THF, 32.4 ml, 32.4 mmol) dropwise. The solution was left to stir at -78 0C for 45 min. prior to dropwise addition of methyl iodide (4.0 ml, 64.9 mmol). The resulting mixture was stirred at -78 0C for a further 30 min. and at then room temperature overnight. The solution was then cooled down to 0 0C then quenched with water, followed by IM HCl. The aqueous phase was extracted with diethyl ether (x3) and the combined organic extracts washed with saturated sodium hydrogen carbonate, followed by brine, dried (MgSO4) and the solvent evaporated. The crude material was purified by flash chromatography using a Biotage SP4 (petroleum ether/ethyl acetate gradient) to give the product as a brown solid (1.0 g, 43%). 1H NMR (400 MHz, CHLOROFORM-rf) δ ppm 1.19 (s, 6 H), 1.53 (s, 9 H), 1.83 (t, .7=6.9 Hz, 2 H), 3.62 - 3.69 (m, 2 H); m/z (ES+APCI)+: 114 [M - CO2'Bu+H]+
Step 3
3, 3-Dimethyl-pyrrolidin-2-one
Figure imgf000087_0001
4 M HCl in dioxane (10 ml) was added to 3,3-Dimethyl-2-oxo-pyrrolidine-l-carboxylic acid tert- butyl ester (990 mg, 4.65 mmol) and the reaction mixture stirred at room temperature for 3 h. The solvents were removed to give the product as an oily brown solid (668 mg, 96%). 1H NMR (400 MHz, DMSCW6) δ ppm 1.03 (s, 6 H), 1.85 (t, /=6.9 Hz, 2 H), 3.15 (t, /=6.4 Hz, 2 H), 5.15 (br s, 1 H); m/z (ES+APCI)+: 114 [M+H]+
Step 4
[3-(3,3-Dimethyl-2-oxo-pyrrolidin-l-yl)-propyl]-carbamic acid tert-butyl ester
Figure imgf000088_0001
Sodium hydride (60% dispersion in mineral oil; 332 mg, 13.9 mmol) was added to anhydrous THF (10 ml), under nitrogen and the mixture cooled down to 0 0C. A solution of 3,3-Dimethyl- pyrrolidin-2-one (668 mg, 4.5 mmol) in anhydrous THF (10 ml) was added dropwise and the reaction mixture stirred at this temperature for 45 min. After this time, a solution of tert-butyl (3- bromopropyl)carbamate (1.28 g, 5.4 mmol) in anhydrous THF (10 ml) was added dropwise and stirring was continued at 0 0C for further 30 min., and then at room temperature overnight. The solution was cooled to 0 0C and then quenched with water, followed by saturated sodium hydrogen carbonate(aq). The aqueous phase was extracted twice with DCM, and the combined organic extracts were washed with brine, dried (MgSO4) and solvent evaporated. Purification by flash chromatography using a Biotage SP4 (ethyl acetate/methanol gradient) gave a clear oil (657 mg, 54%). 1H NMR (400 MHz, DMSCW6) δ ppm 1.05 (s, 6 H), 1.41 (s, 9 H), 1.53 - 1.62 (m, 2 H), 1.82 (t, /=6.6 Hz, 2 H), 2.87 - 2.94 (m, 2 H), 3.18 (t, /=7.1 Hz, 2 H), 3.28 (t, /=6.6 Hz, 2 H), 6.70 (m, IH); m/z (ES+APCI)+: 171 [M - CO2 1BiH-H]+.
Step 5 l-(3-Amino-propyl)-3,3-dimethyl-pyrrolidin-2-one hydrochloride salt
4 M HCl in dioxane (5 ml) was added to [3-(3,3-Dimethyl-2-oxo-pyrrolidin-l-yl)-propyl]-carbamic acid tert-butyl ester (654 mg, 2.4 mmol) and the reaction mixture stirred at room temperature for 1 h. The solvents were removed to give the title compound as a clear oil (645 mg). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.05 (s, 6 H), 1.75 - 1.88 (m, 4 H), 2.68 - 2.78 (m, 2 H), 3.24 - 3.34 (m, 4
H), 7.85 (br s, 2 H); m/z (ES+ APCI)+: 171 [M+H]+
Intermediate 37 l-[3-(2-Chloro-5-cyclopropyl-pyrimidin-4-ylamino)-propyl]-3,3-dimethyl-pyrrolidin-2-one
Figure imgf000089_0001
Intermediate 1 (117 mg, 0.6 mmol), Intermediate 36 (128 mg, 0.6 mmol) and N,N- diisopropylethylamine (330 μl, 1.9 mmol) were mixed together in isopropyl alcohol (5 ml) at 0 0C, stirred for 30 min., then warmed up to room temperature, stirred for 10 min., then warmed up to 55 0C overnight. The reaction mixture was cooled down and partitioned between water and ethyl acetate, extracted twice with ethyl acetate, and the combined organic extracts were washed with brine, dried (MgSO4) and solvents evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (petroleum ether/ethyl acetate gradient) to give the product as a colourless oil (86 mg, 43%). 1H ΝMR (400 MHz, DMSO-J6) δ ppm 0.54 - 0.68 (m, 2 H), 0.89 - 0.96 (m, 2 H), 1.08 (s, 6 H), 1.51 - 1.59 (m, 1 H), 1.72 - 1.81 (m, 2 H), 1.86 (t, J=6.9 Hz, 2 H), 3.28 (t, 7=6.6 Hz, 2 H), 3.32 - 3.37 (m, 4 H), 7.49 - 7.55 (m, 1 H), 7.74 (s, 1 H); ); m/z (ES+APCI)+: 323 [M+H]+
Intermediate 38 1 -(3-Amino-propyl)-3-trifluoromethyl-l H-pyridin-2-one. hydrochloride salt.
Figure imgf000089_0002
Step l ft-β-Oxo-S-trifluoromethyl^H-pyridin-l-ylJ-propylJ-carbamic acid tert-butyl ester
Figure imgf000090_0001
Sodium hydride (60% dispersion in mineral oil, 88 mg, 3.7 mmol) was added to 1 :4 anhydrous DMF/l,2-dimethoxyethane (25 ml) under nitrogen. The mixture was cooled down to 0 0C, then a solution of 2-hydroxy-3-(trifluoromethyl) pyridine (500 mg, 3.1 mmol) in dry DMF (5 ml) was added dropwise and the resulting mixture was stirred for a further 10 min. Lithium bromide (534 mg, 6.1 mmol) was added and the resulting mixture was stirred for 15 min prior to dropwise addition of tert-buty\ (3-bromopropyl)carbamate (876 mg, 3.7 mmol) in DMF (5 ml). After a further 10 min., the reaction mixture was allowed to warm to room temperature and stirring was continued for 3 days. The solution was cooled down to 0 0C, quenched with water and the aqueous phase extracted with ethyl acetate (x3). The combined organic extracts were washed with water followed by brine, dried (MgSO4) and solvents evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (DCM/methanol gradient) to give a yellow oil (111 mg, 11%). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.35 - 1.52 (m, 9 H), 1.76 - 1.88 (m, 2 H), 2.94 - 3.02 (m, 2 H), 3.99 (t, 7=7.1 Hz, 2 H), 6.42 (t, 7=6.9 Hz, 1 H), 6.95 (t, J=5.7 Hz, 1 H), 7.97 (d, 7=7.3 Hz, 1 H), 8.10 (d, 7=6.9 Hz, 1 H); m/z (ES+APCI)+: 221 [M - CO2'Bu+H]+
Step 2 l-(3-Amino-propyl)-3-trifluoromethyl-lH-pyridin-2-one. Hydrochloride salt.
4 M HCl in dioxane (4 ml) was added to [3-(2-Oxo-3-trifluoromethyl-2H-pyridin-l-yl)-propyl]- carbamic acid tert-b\Λy\ ester (107 mg, 0.3 mmol) and the reaction mixture was stirred at room temperature for 3 h. The solvents were removed to give the product as a brown oil (110 mg). 1H
NMR (400 MHz, DMS(W6) δ ppm 1.90 - 2.12 (m, 2 H), 2.78 - 2.87 (m, 2 H), 4.09 (t, 7=7.1 Hz, 2
H), 6.46 (t, 7=6.9 Hz, 1 H), 7.88 - 8.07 (m, 4 H), 8.15 (d, 7=6.9 Hz, 1 H); m/z (ES+APCI)+: 221
[M+H]+
Intermediate 39 l-[3-(2-Chloro-5-cyclopropyl-pyrimidin-4-ylaτnino)-propyl]-3-trifluoromethyl-lH-pyridin-2-one
Figure imgf000091_0001
Intermediate 1 (79 mg, 0.4 mmol), Intermediate 38 (107 mg, 0.4 mmol) and NN- diisopropylethylamine (222 μl, 1.3 mmol) were mixed together in isopropyl alcohol (2 ml) at 0 °C, stirred for 30 min., warmed to room temperature and stirred for 15 min. and then warmed to 55 °C and stirred overnight. The reaction mixture was cooled down and partitioned between water and ethyl acetate and the aqueous phase extracted twice with ethyl acetate, the combined organic extracts washed with brine, dried (MgSO4) and the solvent was evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (petroleum ether/ethyl acetate gradient) to give a white solid (47 mg, 30%). 1H ΝMR (400 MHz, DMSO-J6) δ ppm 0.53 - 0.66 (m, 2 H), 0.88 - 0.95 (m, 2 H), 1.51 - 1.58 (m, 1 H), 1.95 - 2.03 (m, 2 H), 3.39 - 3.48 (m, 2 H), 4.01 - 4.09 (m, 2 H), 6.42 (t, /=6.9 Hz, 1 H), 7.53 (t, /=5.7 Hz, 1 H), 7.74 (s, 1 H), 7.97 (d, /=6.0 Hz, 1 H), 8.11 - 8.15 (m, 1 H); m/z (ES+ APCI)+: 373 [M+H]+
Intermediate 40
5-Cyclopropyl-N4-(3-methylamino-propyl)-N2-(6-morpholin-4-yl-pyridin-3-yl)-pyrimidine-2,4- diamine hydrochloride salt
Figure imgf000091_0002
Step !
{3-[5-Cyclopropyl-2-(6-morpholin-4-yl-pyridin-3-ylamino)ψyrimidin-4-ylamino]-propyl}-methyl- carbamic acid tert-butyl ester
Figure imgf000092_0001
To a stirred solution of Intermediate 28 (194 mg, 0.6 mmol) in 14:1 acetonitrile/water was added 6- morpholinopyridin-3-amine (133 mg, 0.7 mmol) followed by 4M HCl in dioxane (207 μl), and the resulting mixture stirred to 50 0C overnight. The solvents were evaporated and the crude material was purified by preparative LCMS (high pH buffer) to give a purple solid (140 mg, 51%). 1H NMR (400 MHz, DMSCW6, 80 0C) δ ppm 0.47 - 0.54 (m, 2 H), 0.84 - 0.90 (m, 2 H), 1.43 (s, 9 H), 1.46 - 1.55 (m, 1 H), 1.81 - 1.88 (m, 2 H), 2.84 (s, 3 H), 3.31 (t, /=6.9 Hz, 2 H), 3.36 - 3.41 (m, 4 H), 3.43 - 3.49 (m, 2 H), 3.73 - 3.77 (m, 4 H), 6.46 - 6.51 (m, 1 H), 6.78 (d, /=9.2 Hz, 1 H), 7.61 (s, 1 H), 7.98 (dd, /=8.7, 2.7 Hz, 1 H), 8.35 (s, 1 H), 8.48 (d, /=2.7 Hz, 1 H); m/z (ES+APCI)+: 484 [M+H]+
Step 2
5-Cyclopropyl-N4-(3-methylamino-propyl)-N2-(6-morpholin-4-yl-pyridin-3-yl)-pyrimidine-2,4- diamine hydrochloride salt
4 M HCl in dioxane (3 ml) was added to {3-[5-Cyclopropyl-2-(6-morpholin-4-yl-pyridin-3- ylamino)-pyrimidin-4-ylamino]-propyl}-methyl-carbamic acid tert-butyl ester (135 mg, 0.3 mmol) and the reaction mixture was stirred at room temperature for 1.5 h. The volatiles were evaporated to give the product as a purple coloured oil (161 mg). 1H NMR (400 MHz, DMSO-rf6, 80 0C) δ ppm 0.54 - 0.68 (m, 2 H), 0.88 - 0.98 (m, 2 H), 1.57 - 1.65 (m, 1 H), 1.94 - 2.03 (m, 2 H), 2.57 (s, 3 H, obscured by DMSO peak), 2.90 - 2.99 (m, 2 H), 3.51 - 3.80 (m, 6 H, obscured by water peak), 3.74 - 3.91 (m, 4 H), 7.19 - 7.26 (m, 1 H), 7.63 (s, 1 H), 7.86 - 7.93 (m, 1 H), 8.42 (s, 1 H), 8.86
(br. s, 1 H), 9.02 (br. s, 1 H), 10.53 (br. s, 1 H); m/z (ES+APCI)+: 384 [M+H]+
Intermediate 41
6-(l-Methyl-lH-pyrazol-4-yl)-pyridin-3-ylamine
Figure imgf000093_0001
To a solution of 3-amino-6-bromopyridine (400 mg, 2.3 mmol) and l-methyl-4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-pyrazole (721 mg, 3.5 mmol) in 15:1 1,2- dimethoxyethane/water (16 ml), was added potassium carbonate (297 mg, 3.5 mmol), followed by Pd(PPh3)4 (267 mg, 0.2 mmol), under nitrogen, and the resulting mixture was heated at 800C for 16 h. The mixture was cooled, partitioned between water and ethyl acetate, and the aqueous phase was extracted with ethyl acetate. The combined organic phases were washed with water followed by brine, dried (MgSO4) and solvents were evaporated. The crude product was purified by flash chromatography using a Biotage SP4 (DCM/methanol gradient) to give a brown solid (45 mg, 11%). 1H NMR (400 MHz, CHLOROFORM-^) δ ppm 3.65 - 3.80 (br s, 2 H), 3.85 (s, 3 H), 6.91 - 6.95 (m, 1 H), 7.20 (d, 7=7.8 Hz, 1 H), 7.74 (s, 1 H), 7.79 (s, 1 H), 7.99 (d, J=2.7 Hz, 1 H); Rf = 0.43 (1:9 MeOH/DCM).
Intermediate 42 Cyclobutanecarboxylic acid {3-[2-(3-bromomethyl-phenylamino)-5-cyclopropyl-pyrimidin-4- ylaminoj-propylj-amide.
Figure imgf000093_0002
To a solution of Example 70 (120 mg, 0.28 mmol) in glacial acetic acid (10 ml) was added 30%
HBr in AcOH (10 ml). The orange solution was allowed to stir at room temperature for 18h. The reaction mixture was poured into diethyl ether (30 ml), partitioned with water (30 ml), basified with 2M sodium hydroxide solution and the layers separated. The aqueous layer was extracted diethyl ether (30 ml) and DCM (30 ml). The organic phases were combined and washed with brine, dried (MgSO4) and evaporated. The crude material was purified by flash chromatography on the Biotage SP4, eluting with 0 to 10% Methanol/DCM gradient. This gave the desired product as a white solid. 1H NMR (400 MHz, DMSO-^6) δ ppm 0.46 - 0.52 (m, 2 H), 0.81 - 0.90 (m, 2 H), 1.44 - 1.53 (m, 1 H), 1.66 - 1.78 (m, 3 H), 1.79 - 1.93 (m, 3 H), 1.94 - 2.03 (m, 2 H), 2.05 - 2.17 (m, 2 H), 2.90 - 3.01 (m, 1 H), 3.10 - 3.18 (m, 2 H), 3.43 - 3.51 (m, 2 H), 6.93 - 7.00 (m, 1 H), 7.07 - 7.17 (m, 1 H), 7.19 - 7.25 (m, 1 H), 7.52 - 7.58 (m, 1 H), 7.62 (s, 1 H), 7.66 - 7.73 (m, 1 H), 7.95 - 8.01 (m, 1 H), 9.14 (br. s, 1 H); m/z (ES+APCI)+: 458/460 [M+H]+.
Intermediate 43 3-(l-Morpholin-4-yl-ethyl)-phenylamine.
Figure imgf000094_0001
Step l l-(l-Chloro-ethyl)-3-nitro-benzene
Figure imgf000094_0002
Thionyl chloride (162ul, 8.9 mmol) was added dropwise to a solution of l-(3-nitro-phenyl)-ethanol (4.4 mmol) in DCM (50 ml) at 0 0C. The reaction was stirred at room temperature for 2 h. Further thionyl chloride (162 μl, 8.9 mmol) was added and the reaction stirred at 35 0C for 24 h. The mixture was evaporated to give the desired product as a yellow oil. NMR (DMSO) shows 70%, together with 3-nitrostyrene impurity. Used in the next step without further purification. Step 2
4-[l-(3-Nitro-phenyl)-ethyl]-morpholine.
Figure imgf000095_0001
To a solution of the crude l-(l-chloro-ethyl)-3-nitro-benzene (700 mg, 3.78 mmol) in DCM (20 ml) was added diisopropylethylamine (1.23 ml, 7.57 mmol) and then morpholine (494 μl, 5.68 mmol). The reaction was stirred at 40 0C for 18 h, further diisopropylethylamine (1.23 ml, 7.57 mmol) and morpholine (659 μl, 7.57 mmol) were added and the reaction continued for another 18 h. The reaction was diluted with water (50 ml) and the pH was adjusted to 8 and then extracted twice with DCM (40 ml). The combined organics were washed with saturated sodium chloride solution, dried (MgSO4) and evaporated. Purification by flash chromatography on the Biotage SP4, eluting with 0 to 50% ethyl acetate/petroleum ether gradient, gave the desired product as a yellow oil (46%).
1H NMR (400 MHz, DMSCW6) δ ppm 1.30 (d, 7=6.4 Hz, 3 H), 2.23 - 2.31 (m, 2 H), 2.38 - 2.47 (m, 2 H), 3.51 - 3.60 (m, 5 H), 7.61 - 7.66 (m, 1 H), 7.77 - 7.81 (m, 1 H), 8.12 (ddd, /=7.8, 2.3, 1.4 Hz, 1 H), 8.14 - 8.16 (m, 1 H); m/z (ES+APCI)+: 237 [M+H]+.
Step 3
3-(l-Morpholin-4-yl-ethyl)-phenylamine.
Iron powder (329 mg, 5.90 mmol) was added to a solution of Step 2 (470 mg, 1.99 mmol) in acetic acid (6.3 ml) and water (3.9 ml). The reaction was stirred at 70 0C for 1 h, cooled to room temperature, partitioned between ethyl acetate and water. The aqueous phase was extracted twice with ethyl acetate (40 ml), the combined organics were dried (MgSO4) and evaporated to give the title compound as a pale brown oil (88%).
1H NMR (400 MHz, DMSO-^6) δ ppm 1.21 (d, 7=6.9 Hz, 3 H), 2.20 - 2.29 (m, 2 H), 2.30 - 2.42 (m, 2 H), 3.07 (q, J=6.4 Hz, 1 H), 3.48 - 3.59 (m, 4 H), 4.98 (br. s, 2 H), 6.37 - 6.44 (m, 2 H), 6.49 -
6.53 (m, 1 H), 6.88 - 6.97 (m, 1 H); m/z (ES+APCI)+: 207 [M+H]+.
Intermediate 44 Cyclobutanecarboxylic acid {3-[2-(3-chloromethyl-phenylamino)-5-cyclopropyl-pyrimidin-4- ylamino]-propyl}-amide. Hydrochloride.
Figure imgf000096_0001
Thionyl chloride (253 μl, 3.5 mmol) was added dropwise to a suspension of Example 70 (300 mg, 0.69 mmol) in DCM (25 ml) at 0 0C. The mixture was then allowed to warm to room temperature and stirred for 18 h producing a white precipitate. The reaction was evaporated to give the desired product as a white solid (100%).
1H NMR (400 MHz, DMSO-^6) δ ppm 0.55 - 0.60 (m, 2 H), 0.88 - 0.94 (m, 2 H), 1.52 - 1.60 (m, 1 H), 1.66 - 1.77 (m, 3 H), 1.79 - 1.91 (m, 1 H), 1.92 - 2.01 (m, 2 H), 2.03 - 2.14 (m, 2 H), 2.90 - 2.99 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.47 - 3.54 (m, 2 H), 4.79 (s, 2 H), 7.20 - 7.24 (m, 1 H), 7.38 - 7.43 (m, 1 H), 7.46 - 7.51 (m, 1 H), 7.65 (s, 1 H), 7.74 - 7.76 (m, 1 H), 7.76 - 7.81 (m, 1 H), 10.53 (br. s, 1 H), 12.10 (br. s, 1 H); m/z (ES+APCI)+: 414 [M+H]+.
Intermediate 45 (3 -Amino-phenyl) -acetonitri Ie.
Figure imgf000096_0002
Iron powder (517 mg, 9.27 mmol) was added to a solution of 3-nitrophenylacetonitrile (500 mg, 3.09 mmol) in acetic acid (9.5 ml) and water (6.6 ml). The reaction was stirred at 40 0C for 24 h, then cooled to room temperature. The mixture was diluted with water (20 ml), adjusted to pH 8 with 2N NaOH and then filtered trough Celite®. The aqueous filtrate was extracted with ethyl acetate, the combined organic phases were dried (MgSO,)) and evaporated. Purification by flash chromatography on the Biotage SP4, eluting with 0 to 60% ethyl acetate / petroleum ether gradient, gave the desired product as a colourless oil (51%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.84 (s, 2 H), 5.22 (br. s, 2 H), 6.40 - 6.44 (m, 1 H), 6.46 - 6.50 (m, 1 H), 6.50 - 6.53 (m, 1 H), 6.96 - 7.03 (m, 1 H); Rf (50% Ethyl acetate/Petroleum ether) = 0.5.
Intermediate 46 4-(5-Nitro-pyrimidin-2-yl)-morpholine
Figure imgf000097_0001
A mixture of DBPEA (1.1 ml, 6.25 mmol) and morpholine (544 μl, 6.25 mmol) in MeCN (5 ml) was added to a stirred solution of 2-chloro-5-nitropyrimidine (1.0 g, 6.25 mmol) in MeCN (15 ml). The mixture was stirred at rt for 3 days. The reaction mixture was concentrated to dryness and diluted with DCM and saturated NaHCθ3 solution (aq). The organic phase was dried and concentrated to dryness to give an orange coloured oily solid (1.25 g, 95 %). 1H NMR (400 MHz, CHLOROFORM-^) δ ppm 3.77 - 3.86 (m, 4 H), 3.97 - 4.06 (m, 4 H), 9.08 (s, 2 H).
Intermediate 47 2-Morpholin-4-yl-pyrimidin-5-ylamine
Figure imgf000097_0002
Prepared analogously to Intermediate 13 using Intermediate 46 to afford an orange coloured solid (1.04 g, 97 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.36 - 3.50 (m, 4 H), 3.57 - 3.68 (m, 4 H), 4.66 (s, 2 H), 7.90 (s, 2 H); m/z (ES+APCI)+ : 181 [M+H]+.
Intermediate 48
4-[2-(5-Nitro-pyridin-2-yloxy)-ethyl]-morpholine
Figure imgf000097_0003
Sodium hydride, 60 % dispersion in oil (252 mg, 6.29 mmol) was added portionwise to a solution of 2-chloro-5-nitropyridine (1.0 g, 6.29 mmol) and 2-morpholin-4-yl-ethanol (785 mg, 5.99 mmol) in DMF (20 ml) with ice-cooling. The ice-cooling was removed after 1 hour and the mixture was stirred at rt overnight. The reaction mixture was added to an ice/water mixture and extracted with
EtOAc (x2). The combined organic extracts were washed with water (x4) and brine (xl), dried and concentrated to dryness. The residue was purified by flash column chromatography on silica gel (50 g) eluting with 40:1 DCM-MeOH to provide a brown solid (980 mg, 62 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.45 (br. s, 4 H), 2.71 (t, J=5.72 Hz, 2 H), 3.49 - 3.61 (m, 4 H), 4.52 (t, /=5.72 Hz, 2 H), 7.04 (d, /=9.16 Hz, 1 H), 8.44 - 8.49 (m, 1 H), 9.08 (d, /=3.66 Hz, 1 H); m/z (ES+APCI)+ : 254 [M+H]+.
Intermediate 49 6-(2-Morpholin-4-yl-ethoxy)-pyridin-3-ylamine
Figure imgf000098_0001
Prepared analogously to Intermediate 13 using Intermediate 48 to give an orange oil (883 mg, 102%). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.36 - 2.48 (m, 4 H), 2.60 (t, /=5.95 Hz, 2 H), 3.51 - 3.59 (m, 4 H), 4.19 (t, /=5.95 Hz, 2 H), 4.73 (s, 2 H), 6.52 (d, /=8.70 Hz, 1 H), 6.96 - 7.00 (m, 1 H), 7.47 (d, /=3.66 Hz, 1 H).
Intermediate 50 4-[2-(4-Nitro-pyrazol-l-yl)-ethyl]-morpholine
Figure imgf000098_0002
N-(2-chloroethyl)morpholine HCl salt (2.1 g, 11.06 mmol) was added portionwise to a stirred mixture of 4-nitro-lH-pyrazole (1.0 g, 8.85 mmol) and KOH (1.24 g, 22.12 mmol) in EtOH (20 ml). The mixture was heated under reflux for 2 h and allowed to cool to rt. After dilution with EtOAc and water the organic phase was washed with brine, dried and concentrated. The residue was purified by flash column chromatography on silica gel (100 g) eluting with 50:1 DCM-MeOH to provide an orange oil (987 mg, 49 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.30 - 2.49 (m, 4 H), 2.73 (t, /=6.18 Hz, 2 H), 3.42 - 3.61 (m, 4 H), 4.30 (t, /=6.18 Hz, 2 H), 8.26 (s, 1 H), 8.88 (s, 1 H); m/z (ES+APCI)+ : 227 [M+H]+. Intermediate 51 l-(2-Morpholin-4-yl-ethyl)-lH-pyrazol-4-ylamine
Figure imgf000099_0001
Prepared analogously to Intermediate 13 using Intermediate 50 to provide a red coloured oil (775 mg, 91 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.30 - 2.44 (m, 4 H), 2.61 (t, J=6.64 Hz, 2 H), 3.47 - 3.59 (m, 4 H), 3.84 (br. s, 2 H), 4.02 (t, J=6.64 Hz, 2 H), 6.88 (s, 1 H), 7.05 (s, 1 H); m/z (ESH-APCI)+ : 197 [M+H]+.
Intermediate 52 l-[2-(4-Nitro-pyrazol-l-yl)-ethyl]-piperidine
Figure imgf000099_0002
Prepared analogously to Intermediate 50 using N-(2-chloroethyl)piperidine HCl salt and 4-nitro- lH-pyrazole to provide the product as an orange coloured solid (1.94 g, 98 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.27 - 1.55 (m, 6 H), 2.25 - 2.48 (m, 4 H), 2.65 (t, 7=6.41 Hz, 2 H), 4.26 (t, J=6.41 Hz, 2 H), 8.24 (s, 1 H), 8.84 (s, 1 H); m/z (ES+APCI)+ : 225 [M+H]+.
Intermediate 53 l-(2-Piperidin-l-yl-ethyl)-lH-pyrazol-4-ylamine
Figure imgf000099_0003
Prepared analogously to Intermediate 13 using Intermediate 52 to afford a red coloured solid (1.64 g, 98 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.19 - 1.41 (m, 2 H), 1.41 - 1.57 (m, 4 H), 2.33 (br. s, 4 H), 2.45 - 2.64 (m, 2 H), 3.84 (br. s, 2 H), 3.90 - 4.08 (m, 2 H), 6.86 (s, 1 H), 7.03 (s, 1 H); m/z (ES+APCI)+ : 195 [M+H]+. Intermediate 54
(l-Cyclobutanecarbonyl-azetidin-3-ylmethyl)-carbamic acid tert-butyl ester
Figure imgf000100_0001
To a solution of azetidin-3-ylmethyl-carbamic acid tert-butyl ester (279 mg, 1.5 mmol) in DCM was added DIPEA (550 μl, 3.15 mmol) followed by cyclobutanecarbonyl chloride (170 μl, 1.5 mmol) dropwise at 0 °C. The reaction was stirred for 6 h at RT, quenched with NaHCO3(aq) and washed with brine. The organic fractions were isolated by phase separation cartridge and concentrated under vacuum to give the desired product. (187 mg, 47%). 1H NMR (400 MHz,
DMSO-ck) δ ppm 1.37 (s, 9 H), 1.66 - 1.82 (m, 1 H), 1.82 - 1.94 (m, 1 H), 1.94 - 2.05 (m, 1 H), 2.05 - 2.16 (m, 3 H), 2.54 - 2.65 (m, 1 H), 2.97 - 3.07 (m, 1 H), 3.05 - 3.13 (m, 1 H), 3.33 (s, 1 H),
3.50 (dd, 7=9.62, 5.50 Hz, 1 H), 3.65 (dd, 7=8.70, 5.50 Hz, 1 H), 3.75 - 3.84 (m, 1 H), 4.00 (t,
7=8.47 Hz, 1 H), 7.04 (t, 7=5.72 Hz, 1 H).
Intermediate 55
(3-Aminomethyl-azetidin-l-yl)-cyclobutyl-methanone
Figure imgf000100_0002
To a solution of Intermediate 54 (187 mg, 0.7 mmol) in MeOH was added 4M HCl in dioxane (1.75 ml, 7 mmol) and stirred overnight at RT. The product was purified on SCX cartridge to give the desired product (66 mg, 57%). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.67 - 1.78 (m, 1 H), 1.81 - 1.94 (m, 1 H), 1.94 - 2.04 (m, 2 H), 2.04 - 2.14 (m, 2 H), 2.63 - 2.69 (m, 1 H), 2.96 - 3.13 (m, 2 H), 3.16 (s, 1 H), 3.48 (dd, 7=9.39, 5.27 Hz, 1 H), 3.67 (dd, 7=8.47, 5.27 Hz, 1 H), 3.78 (t, 7=8.93 Hz, 1 H), 3.99 (t, 7=8.24 Hz, 1 H); m/z (ES+APCI)+ : 169 [M+H]+.
Intermediate 56 {3-[(2-Chloro-5-cyclopropylφyrimidin-4-ylamino)-methyl]-azetidin-l-yl}-cyclobutyl-methanone
Figure imgf000101_0001
To a solution of Intermediate 1 (75 mg, 0.4 mmol) and DIPEA (155 mg, 1.2 mmol) in IPA was added Intermediate 55 (66 mg, 0.4 mmol) portionwise at 0 0C and stirred overnight at RT. The reaction was concentrated, taken up in DCM, washed with water and brine and purified by Biotage SP4 (ethyl acetate / petroleum ether gradient) to give the desired product as a colourless oil (37 mg, 29%). 1H NMR (400 MHz, CHLOROFORM-rf) δ ppm 0.47 - 0.59 (m, 2 H), 0.88 - 1.02 (m, 2 H), 1.35 - 1.46 (m, 1 H), 1.79 - 1.92 (m, 1 H), 1.92 - 2.00 (m, 1 H), 1.99 - 2.11 (m, 1 H), 2.09 - 2.23 (m, 2 H), 2.24 - 2.42 (m, 3 H), 3.09 - 3.32 (m, 2 H), 3.42 - 3.54 (m, 1 H), 3.54 - 3.73 (m, 1 H), 3.78 - 3.90 (m, 1 H), 4.64 - 4.83 (m, 1 H), 5.43 (d, ./=6.41 Hz, 1 H), 7.84 (d, 7=6.41 Hz, 1 H).
Intermediate 57
4-{4-[3-(Cyclobutamcarbonyl-amino)-propylamino]-5-cyclopropyl-pyrimidin-2-ylamino}-benzoic acid
Figure imgf000101_0002
A solution of Intermediate 8 (200 mg, 0.65 mmol), 4-aminobenzoic acid (267 mg, 1.95 mmol) and glacial AcOH (12 mg, 0.2 mmol) in π-butanol was heated at 150 0C in the microwave for 35 min. The resulting precipitate was filtered and dried in a drying pistol to give the desired product (221 mg, 83%). 1H NMR (400 MHz, DMSO-ύfe) δ ppm 0.52 - 0.64 (m, 2 H), 0.87 - 0.98 (m, 2 H), 1.49 -
1.61 (m, 1 H), 1.67 - 1.78 (m, 4 H), 1.78 - 1.88 (m, 1 H), 1.89 - 2.00 (m, 2 H), 2.02 - 2.14 (m, 2 H), 2.94 (t, /=8.01 Hz, 1 H), 3.08 - 3.18 (m, 2 H), 3.51 (q, ./=6.72 Hz, 2 H), 7.67 (s, 1 H), 7.73 (d, J=8.70 Hz, 2 H), 7.76 (t, J=5.72 Hz, 1 H), 7.98 (d, 7=9.16 Hz, 2 H), 8.62 (br. s, 1 H), 10.56 (br. s, 1 H); m/z (ES+APCI)+ : 410 [M+H]+.
Intermediate 58
2-Methyl-4-nitro-benzoic acid methyl ester
Figure imgf000102_0001
A solution of 2-methyl-4-nitro-benzoic acid (10 g, 55 mmol), MeI (3.8 ml, 61 mmol) and K2CO3 (11.4 g, 83 mmol) in DMF was stirred for 3 h at RT after which the reaction was poured onto water and extracted in EtOAc. The organic fractions were washed with water and brine, dried over Na2SO4 and concentrated to give the desired product as an orange solid (9.57g, 89%). 1H NMR (400 MHz, DMSO-^6) δ ppm 2.60 (s, 3 H), 3.88 (s, 3 H), 8.00 (d, J=8.70 Hz, 1 H), 8.10 - 8.16 (m, 1 H), 8.21 (d, J=2.29 Hz, I H).
Intermediate 59
2-Bromomethyl-4-nitro-benzoic acid methyl ester
Figure imgf000102_0002
A solution of Intermediate 58 (9.57 g, 49 mmol), NBS (9.6g, 54 mmol), benzoyl peroxide (121 mg,
0.5 mmol) in carbon tetrachloride was heated at 85°C for 18 h. The reaction was concentrated under vaccum, analysed by TLC and 1H NMR and found to have some remaining starting material. The residues were added to CCl4 (40 ml) with NBS (4.4 g) and benzoyl peroxide (60 mg) then stirred at 85°C for 3h. The reaction was concentrated, taken up in DCM then washed with aqueous NaHCO3 and brine. The organic phase was dried over Na2SO4 and concentrated under vacuum. The residues were purified by flash chromatography using a Biotage SP4. The desired product was isolated as an orange solid (6.58 g, 49%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.93 (s, 3 H), 5.10 (s, 2 H), 8.08 (d, 7=8.24 Hz, 1 H), 8.25 - 8.30 (m, 1 H), 8.51 (d, /=2.29 Hz, 1 H).
Intermediate 60
2-Methyl-5-nitro-2,3-dihydro-isoindol-l-one
Figure imgf000103_0001
A solution of Intermediate 59 (1 g, 3.7 mmol), methylamine hydrochloride (124 mg, 4 mmol) and
Et3N (404 mg, 4 mmol) were refluxed in MeOH for 24 h under N2. During this time an additional 0.5eq of methylamine hydrochloride was added, twice, to compensate for the volatility of methylamine. The mixture was diluted with EtOAc, washed sequentially with IM HCl and brine., and the organic phase was dried over Na2SO4 and concentrated under vacuum. The residues were purified by silica gel chromatography, eluting first with 20% EtOAc in petroleum ether and then with 30% MeOH in DCM. Further purification by preparative LCMS gave the title compound (62 mg, 9%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.12 (s, 3 H), 4.60 (s, 2 H), 7.90 (d, J=8.24 Hz, 1 H), 8.26 - 8.36 (m, 1 H), 8.51 (s, 1 H); m/z (ES+APCI)+ : 193 [M+H]+.
Intermediate 61
2-(2-Hydroxy-ethyl)-5-nitro-2,3-dihydro-isoindol-l-one
Figure imgf000103_0002
A solution of Intermediate 59 (1 g, 3.7 mmol), 2-hydroxy ethylamine (242 μl, 4 mmol) and Et3N (404 mg, 4 mmol) were refluxed in MeOH for 24 h under N2. The mixture was diluted in DCM, washed with IM HCl and brine. The organic fractions were dried over Na2SO4 and concentrated under vacuum. The residues were purified by preparative LCMS to give the title compound (114 mg, 14%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.55 - 3.69 (m, 4 H), 4.68 (s, 2 H), 4.86 - 4.95 (m, 1 H), 7.91 (d, /=8.24 Hz, 1 H), 8.33 (dd, /=8.47, 2.06 Hz, 1 H), 8.52 (s, 1 H); m/z (ES+APCI)+ : 223 [M+H]+.
Intermediate 62
5-Amino-2-methyl-2,3-dihydro-isoindol-l-one
Figure imgf000104_0001
A mixture of Intermediate 60 (68 mg), Fe (180 mg), and HCl (53 μl), in EtOH (3 ml), was refluxed at 950C under N2 for 3 h. The reaction was basified with a few drops of 2M NH3 in MeOH then concentrated under vacuum. The residues were taken up in 20% MeOH in DCM, filtered twice through a plug of silica gel, and the filtrate was concentrated under vacuum to give the desired product (58 mg, 99%). 1H NMR (400 MHz, DMSO-^6) δ ppm 2.96 (s, 3 H), 4.24 (s, 2 H), 5.75 (br. s, 2 H), 6.54 - 6.64 (m, 2 H), 7.27 (d, /=7.78 Hz, 1 H); m/z (ES+APCI)+ : 163 [M+H]+.
Intermediate 63 5-Amino-2-(2-hydroxy-ethyl)-2, 3-dihydro-isoindol-l-one
Figure imgf000104_0002
Prepared analogously to Intermediate 62 from Intermediate 61 (114 mg, 0.5 mmol) to give the title compound (87 mg, 89%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.49 - 3.61 (m, 2 H), 4.32 (s, 2 H), 4.78 (t, /=5.27 Hz, 1 H), 5.69 (s, 2 H), 6.51 - 6.64 (m, 2 H), 7.16 (br. s, 2 H), 7.28 (d, /=8.24 Hz, 1 H): m/z (ES+APCI)+ : 193 [M+H]+. Intermediate 64
6-Oxazol-5-yl-pyridin-3-ylamine
Figure imgf000105_0001
5-Nitro-2-oxazol-5-yl-pyridine
Figure imgf000105_0002
To a solution of 5-nitro-pyridine-2-carbaldehyde (650 mg, 4.3 mmol) in MeOH (30 ml) was added tosyl isocyanide (840 mg, 4.3 mmol) followed by K2CO3 and heated with stirring at 7O0C for 2.5 h. The mixture was poured onto ice water and extracted with EtOAc then DCM. The organic components were washed with aqueous HCl (IM) and brine before drying over MgSθ4. The combined organic fractions were concentrated under vacuum to give the desired product as a yellow solid (548 mg, 67 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 8.04 (d, /=8.70 Hz, 1 H), 8.10 (s, 1 H), 8.68 - 8.74 (m, 2 H), 9.42 (d, ./=2.75 Hz, 1 H).
Step 2
6-Oxazol-5-yl-pyridin-3-ylamine
To a solution of 5-nitro-2-oxazol-5-yl-pyridine (545 mg, 2.8 mmol) in EtOH (20 ml) was added
Pd/C (30 mg, 0.28 mmol). The flask was evacuated and filled with N2 three times then evacuated and filled with H2 three times before stirring overnight at RT under H2. The mixture was filtered through Celite under N2, concentrated under reduced pressure and separated on Biotage SP4 (DCM / MeOH gradient) to give the desired product as a yellow solid (106 mg, 24 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 7.27 (dd, J=8.70, 2.75 Hz, 1 H), 7.49 - 7.57 (m, 1 H), 7.59 (d, 7=8.24 Hz, 1 H), 8.17 (d, J=2.75 Hz, 1 H), 8.39 - 8.45 (m, 1 H), 8.70 (d, 7=1.83 Hz, 1 H), 8.83 (s, 1 H); m/z (ES+APCI)+ : 162 [M+H]+. Intermediate 65
Imidazo[l,2-a]pyridin-6-ylamine
Figure imgf000106_0001
To a solution of 6-nitro-imidazo[l,2-a]pyridine (500 mg, 3 mmol) in EtOH (20 ml) was added Pd/C (30 mg, 0.3 mmol). The flask was evacuated and filled with N2 three times then evacuated and filled with H2 three times before stirring overnight at RT under H2. The mixture was filtered through celite under N2, concentrated under reduced pressure and run through a short silica column. The eluant was concentrated under vacuum to give the desired product as a dark green solid (295 mg, 74 %). 1H NMR (400 MHz, DMSO-(Z6) δ ppm 4.87 (br. s, 2 H), 6.78 - 6.83 (m, 1 H), 7.27 - 7.36 (m, 2 H), 7.67 (dd, 1 H), 7.69 (s, 1 H); m/z (ES+APCI)+ : 134 [M+H]+.
Intermediate 66 N -(Tetrahydro-pyran-4-ylmethyl)-pyridine-2, 5-diamine
Figure imgf000106_0002
Step l (5-Nitro-pyridin-2-yl)-(tetrahydro-pyran-4-ylmethyl)-amine
Figure imgf000106_0003
2-Chloro-5-nitro-pyridine (200 mg, 1.3 mmol), 4-aminomethyltetrahydropyran (145 mg, 1.3 mmol) and DIPEA (220 μl, 1.3 mmol) were combined in MeCN (5 ml) and stirred overnight at RT. The resulting precipitate was removed and the filtrate concentrated under vacuum. The filtrate residues were purified on Biotage SP4 (ethyl acetate / petroleum ether gradient) to give the product as a bright green solid (245 mg, 82 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 1.10 - 1.32 (m, 2 H), 1.60 (d, J=13.28 Hz, 2 H), 1.80 (m, 1 H), 3.20 - 3.33 (m, 4 H), 3.84 (dd, J=I 1.22, 2.52 Hz, 2 H), 6.57 (d, J=9.16 Hz, 1 H), 8.08 (d, J=8.24 Hz, 1 H), 8.21 (t, J=5.04 Hz, 1 H), 8.90 (d, 1 H); m/z (ES+APCI)+ : 238 [M+H]+.
Step 2
N -(Tetrahydro-pyran-4-ylmethyl) -pyridine-2, 5 -diamine
To a solution of (5-Nitro-pyridin-2-yl)-(tetrahydro-pyran-4-ylmethyl)-amine (240 mg, 1 mmol) in EtOH (20 ml) was added Pd/C (15 mg, 0.15 mmol). The flask was evacuated and filled with N2 three times then evacuated and filled with H2 three times before stirring overnight at RT under H2. The mixture was filtered through Celite under N2, concentrated under reduced pressure to give the desired product as a red oil ( 104 mg, 50 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.05 - 1.26 (m, 2 H), 1.61 (d, J=12.82 Hz, 2 H), 1.71 (m, 1 H), 2.98 (t, 7=6.41 Hz, 2 H), 3.17 - 3.30 (m, 2 H), 3.83 (dd, J=I 1.22, 2.52 Hz, 2 H), 4.24 (s, 2 H), 5.63 (t, J=5.72 Hz, 1 H), 6.29 (d, J=8.70 Hz, 1 H), 6.79 (dd, J=8.70, 2.75 Hz, 1 H), 7.42 (d, 1 H); m/z (ES+APCI)+ : 208 [M+H]+.
Intermediate 67 6-(l-Cyclopropyl-piperidin-4-yloxy)-pyridin-3-ylamine
Figure imgf000107_0001
Step l
1 -Cyclopropyl-piperidin-4-ol
Figure imgf000108_0001
l-Cyclopropyl-piperidin-4-one (500 mg, 3.6 mmol) and NaBH4 (73 mg, 1.9 mmol) were combined in EtOH and Stirred at RT for 3 h. The reaction was quenched with H2O and extracted with DCM and 2M NaOH(aq). The organic fractions were dried over MgSO4 and concentrated under vacuum to give the desired product as a colourless oil (253 mg, 50 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.17 - 0.29 (m, 2 H), 0.32 - 0.43 (m, 2 H), 1.19 - 1.37 (m, 2 H), 1.47 - 1.58 (m, 1 H), 1.59 - 1.72 (m, 2 H), 2.09 - 2.29 (m, 2 H), 2.70 - 2.86 (m, 2 H), 3.43 (dt, J=8.93, 4.69 Hz, 1 H), 4.54 (d, 1 H); m/z (ES+ APCI)+ : 142 [M+H]+.
Step 2
2-(l-Cyclopropyl-piperidin-4-yloxy)-5-nitro-pyridine
Figure imgf000108_0002
To a solution of l-cyclopropyl-piperidin-4-ol and 5-nitro-2-chloro pyridine in DMF was added NaH portionwise with ice cooling. The reaction was stirred at O0C for 1 h then stirred overnight at RT. The mixture was added to ice water and extracted with EtOAc. The organic extracts were washed with water and brine then dried over MgSO4 and concentrated under vacuum. The residues were purified by Biotage SP4 (ethyl acetate / petroleum ether gradient) to give the desired product as a yellow solid (472 mg, 19 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.23 - 0.36 (m, 2 H), 0.36 - 0.47 (m, 2 H), 1.54 - 1.73 (m, 3 H), 1.90 - 2.04 (m, 2 H), 2.42 (t, 7=9.39 Hz, 2 H), 2.78 - 2.90 (m, 2 H), 5.14 (dt, 7=8.47, 4.46 Hz, 1 H), 7.00 (d, 7=9.16 Hz, 1 H), 8.45 (dd, 7=9.16, 3.21 Hz, 1 H), 9.08 (d, 1 H).
Step 3 6-(l-Cyclopropyl-piperidin-4-yloxy)-pyridin-3-ylamine To a solution of 2-(l-cyclopropyl-ρiperidin-4-yloxy)-5-nitro-pyridine (240 mg, 1 mmol) in EtOH
(20 ml) was added Pd/C (15 mg, 0.15 mmol). The flask was evacuated and filled with N2 three times then evacuated and filled with H2 three times before stirring overnight at RT under H2. The mixture was filtered through celite under N2, concentrated under reduced pressure to give the desired product as a brown oil (104 mg, 50 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.23 - 0.31 (m, 2 H), 0.36 - 0.45 (m, 2 H), 1.43 - 1.55 (m, 2 H), 1.59 (tt, 7=6.75, 3.55 Hz, 1 H), 1.80 - 1.92 (m, 2 H), 2.28 - 2.41 (m, 2 H), 2.75 - 2.87 (m, 2 H), 4.67 - 4.81 (m, 3 H), 6.49 (d, 7=8.70 Hz, 1 H), 6.97 (dd, /=8.70, 2.75 Hz, 1 H), 7.47 (d, 1 H); m/z (ES+APCI)+ : 233 [M+H]+.
Intermediate 68
[J, 2, 4]Triazolo[l, 5-a]pyridin-6-ylamine
Figure imgf000109_0001
Step 1
Benzhydrylidene-[1 , 2, 4]triazolo[l, 5-a]pyridin-6-yl-amine
Figure imgf000109_0002
6-Bromo-[l,2,4]triazolo[l,5-a]pyridine (300 mg, 1.5 mmol), bezophenone imine (326 mg, 1.8 mmol), Pd2(dba)3 (7 mg, 0.008 mmol), BINAP (14 mg, 0.02 mmol) and sodium terf-butoxide (202 mg, 2.1 mmol) were combined in toluene and heated at 80 0C overnight. The mixture was concentrated under reduced pressure and purified by Biotage SP4 (ethyl acetate / petroleum ether gradient) to give the desired product as a yellow crystalline solid (310 mg, 69 %). 1H NMR (400 MHz, DMSO-d6) δ ppm 7.21 (dd, J=9.39, 2.06 Hz, 1 H), 7.24 - 7.29 (m, 2 H), 7.32 - 7.40 (m, 3 H), 7.47 - 7.54 (m, 2 H), 7.55 - 7.62 (m, 1 H), 7.66 (d, J=8.70 Hz, 1 H), 7.68 - 7.74 (m, 2 H), 8.35 (s, 1 H), 8.41 (d, J=I.83 Hz, 1 H); m/z (ES+APCI)+ : 299 [M+H]+.
Step 2
[1, 2, 4]Triazolo[l, 5-a]pyridin-6-ylamine
Figure imgf000110_0001
Benzhydrylidene-[l,2,4]triazolo[l,5-a]pyridin-6-yl-amine (260 mg, 0.87 mmol), hydroxylamine hydrochloride (110 mg, 1.6 mmol) and NaOAc (172 mg, 2.1 mmol) were combined in MeOH and stirred at RT overnight The mixture was concentrated, taken up in DCM, washed with 0.1 M NaOH(aq) and dried by phase separator cartridge. The eluant was concentrated under vacuum, taken up in MeOH and run through an SCX cartridge. The filtrate was discarded and the product eluted with 2M NH3 in methanol, evaporating to give a blue/green crystalline solid (47 mg, 40%). 1H NMR (400 MHz, DMSO-J6) δ ppm 5.26 (br. s, 2 H), 7.11 - 7.26 (m, 1 H), 7.56 (d, J=I 0.07 Hz, 1 H), 8.02 (d, J=2.75 Hz, 1 H), 8.17 (s, 1 H); m/z (ES+APCI)+ : 135 [M+H]+.
Intermediate 69
3-(l-Methyl-l-morpholin-4-yl-ethyl)-phenylamine
Figure imgf000110_0002
Stepl 4-[l-(3-Bromo-phenyl)-l-methyl-ethyl]-morpholine
Figure imgf000110_0003
A mixture of l-(3-bromo-phenyl)-l-methyl-ethylamine (742 mg, 3.47 mmol), 1 -bromo-2-(2- bromo-ethoxy)-ethane (522 μl, 4.16 mmol) and potassium carbonate (957 mg, 6.93 mmol) in ethanol (15 ml) was stirred and heated under reflux for 40 hours. The mixture was allowed to cool to it and diluted with EtOAc and water. The organic phase was washed with brine, dried and concentrated. The crude product was purified by flash column chromatography on silica gel (40 g) eluting with 7:1 petroleum etheπEtOAc to give the product as a colourless oil (585 mg, 59%). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.25 (s, 6 H), 2.30 - 2.41 (m, 4 H), 3.40 - 3.62 (m, 4 H), 7.26 - 7.31 (m, 1 H), 7.38 - 7.42 (m, 1 H), 7.50 (d, 7=7.79 Hz, 1 H), 7.58 - 7.78 (m, 1 H). Step 2
Benzhydrylidene-[3-(l-methyl-l-morpholin-4-yl-ethyl)-phenyl]-amine
Figure imgf000111_0001
A mixture of 4-[l-(3-bromo-phenyl)-l-methyl-ethyl]-morpholine (585 mg, 2.06 mmol), benzophenone imine (447 mg, 2.47 mmol), sodium ter/-butoxide (277 mg, 2.88 mmol), BINAP (49 mg, 0.078 mmol) and Pd2(dba)3 (25 mg, 0.027 mmol) in toluene (15 ml) was placed in a round bottomed flask. The mixture was degassed, placed under an atmosphere of nitrogen and stirred and heated at 800C overnight. On cooling to rt the mixture was diluted with EtOAc and water. The organic phase was washed with brine, dried and concentrated. The crude product was purified by flash column chromatography on silica gel (50 g) eluting with 3:1 petroleum etheπEtOAc to give the product as a yellow/green coloured oil (730 mg, 92%). 1H NMR (400 MHz, DMSO-Of6) δ ppm 1.14 (s, 6 H), 2.09 - 2.17 (m, 4 H), 3.39 - 3.48 (m, 4 H), 6.63 - 6.67 (m, 2 H), 6.98 (d, /=7.79 Hz, 1 H), 7.06 - 7.14 (m, 3 H), 7.25 - 7.29 (m, 3 H), 7.39 - 7.57 (m, 3 H), 7.66 (d, J=6.87 Hz, 2 H).
Step 3 3-(l-Methyl-l-morpholin-4-yl-ethyl)-phenylamine
A mixture of benzhydrylidene-[3-(l-methyl-l-moφholin-4-yl-ethyl)-phenyl]-amine (697 mg, 1.82 mmol), sodium acetate (357 mg, 4.36 mmol) and hydroxylamine hydrochloride (225 mg, 3.27 mmol) in methanol (20 ml) was stirred at rt for 2 hours. The mixture was concentrated to a small volume and diluted with DCM and 0.1M NaOH (aq). The organic phase was dried and concentrated. The crude residue was dissolved in methanol and passed through a SCX cartridge.
The product was eluted with 2M ammonia in methanol to provide a yellow solid after concentration of the eluent (314 mg, 79%). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.24 (s, 6 H),
2.31 - 2.40 (m, 4 H)1 3.49 - 3.58 (m, 4 H), 4.95 (s, 2 H), 6.38 (d, ./=9.16 Hz, 1 H), 6.63 (d, 7=7.78
Hz, 1 H), 6.74 (s, 1 H), 6.93 (t, 7=7.79 Hz, 1 H).
Intermediate 70 l-(Tetrahydro-pyran-4-yl)-lH-pyrazol-4-ylamine
Figure imgf000112_0001
Step l 4-Nitro-l -(tetrahydro-pyran-4-yl)-l H-pyrazole
Figure imgf000112_0002
A solution of di-tert-butyl azodicarboxylate (2.6 g, 11.5 mmol) in THF (5 ml) was added to a stirred solution of tetrahydro-pyran-4-ol (903 mg, 8.85 mmol), 4-nitro-l H-pyrazole (1.0 g, 8.85 mmol) and triphenylphosphine (2.8 g, 10.62 mmol) in THF (35 ml) at rt. The mixture was stirred at rt overnight and subsequently concentrated to dryness. The crude product was purified by flash column chromatography on silica gel (250 g) eluting with 4:1 to 1:1 petroleum etheπEtOAc to give the product as a white solid (1.59 g, 91%). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.90 - 2.04 (m, 4 H), 3.44 (td, J=I 1.33, 3.43 Hz, 2 H), 3.92 - 3.99 (m, 2 H), 4.46 - 4.55 (m, 1 H), 8.29 (s, 1 H), 8.96 (S3 1 H).
Step 2 l-(Tetrahydro-pyran-4-yl)-lH-pyrazol-4-ylamine
Figure imgf000112_0003
A mixture of 4-nitro-l-(tetrahydro-pyran-4-yl)-lH-pyrazole (1.59 g, 8.07 mmol) and 10% palladium on charcoal in ethanol (30 ml) was stirred at it overnight under a balloon of hydrogen. The mixture was filtered through Celite and the filtrate was concentrated to give the product as a purple coloured solid (1.33 g, 98%). 1H NMR (400 MHz, DMSO-^6) δ ppm 1.76 - 1.94 (m, 4 H), 3.29 - 3.48 (m, 2 H), 3.80 (br. s, 2 H), 3.84 - 4.00 (m, 2 H), 4.16 (tt, ./=10.42, 5.38 Hz, 1 H), 6.90 (s, 1 H), 7.06 (s, 1 H); m/z (ES+APCI)+: 168 [M+H]+.
Intermediate 71 and Intermediate 72 1 -Cyclopr opylmethyl-6-nitr o- lH-indazole (Intermediate 71)
Figure imgf000113_0001
2-Cyclopropylmethyl-6-nitro-2H-indazole (Intermediate 72)
Figure imgf000113_0002
A mixture of 6-nitroindazole (2.0 g, 12.3 mmol), cyclopropylmethyl bromide (1.19 ml, 12.3 mmol) and potassium carbonate (3.39 g, 24.5 mmol) in DMF were stirred at 35°C overnight. The mixture was cooled to rt and diluted with EtOAc and water. The organic phase was washed with water (x3), washed with brine (xl), dried and concentrated. The crude product was purified by flash column chromatography on silica gel (125 g) eluting with 4: 1 to 2:1 petroleum ether:EtOAc to provide to isomeric products. The first eluting product was l-cyclopropylmethyl-6-nitro-lH-indazole (Intermediate 71) obtained as a yellow solid (1.14 g, 43%). 1H NMR (400 MHz, CHLOROFORM- d) δ ppm 0.42 - 0.54 (m, 2 H), 0.59 - 0.72 (m, 2 H), 1.33 - 1.44 (m, 1 H), 4.37 (d, 7=6.87 Hz, 2 H), 7.86 (d, /=8.24 Hz, 1 H), 7.99 - 8.06 (m, 1 H), 8.14 (s, 1 H), 8.43 (s, 1 H).
The second eluting product was 2-cyclopropylmethyl-6-nitro-2H-indazole (Intermediate 72) obtained as an orange solid (1.25 g, 47%). 1H NMR (400 MHz, CHLOROFORM-^) δ ppm 0.45 - 0.61 (m, 2 H), 0.63 - 0.82 (m, 2 H), 1.43 - 1.53 (m, 1 H), 4.37 (d, J=7.33 Hz, 2 H), 7.79 (d, /=9.16 Hz, 1 H), 7.92 (dd, 7=9.16, 1.83 Hz, 1 H), 8.19 (s, 1 H), 8.72 (s, 1 H); m/z (ES+APCI)+: 218
[M+H]+.
Intermediate 73 1 -Cyclopropylmethyl-lH-indazol-ό-ylamine
Figure imgf000114_0001
Prepared analogously to Intermediate 70 (Step 2) using l-cyclopropylmethyl-ό-nitro-lH-indazole (Intermediate 71). The product was obtained as a red oil (1.0 g, 102%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.31 - 0.40 (m, 2 H), 0.40 - 0.50 (m, 2 H), 1.15 - 1.25 (m, 1 H), 4.06 (d, 7=6.87 Hz, 2 H), 5.30 (s, 2 H), 6.46 - 6.52 (m, 2 H), 7.35 (d, 7=7.78 Hz, 1 H), 7.69 (s, 1 H); m/z (ES+APCI)+: 188 [M+H]+.
Intermediate 74
2-Cyclopropylmethyl-2H-indazol-6-ylamine
Figure imgf000114_0002
Prepared analogously to Intermediate 70 (Step 2) Intermediate 72. The product was obtained as a brown oil (850 mg, 79%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.31 - 0.45 (m, 2 H), 0.45 - 0.59 (m, 2 H), 1.26 - 1.37 (m, 1 H), 4.09 (d, 7=7.33 Hz, 2 H), 5.02 (s, 2 H), 6.44 - 6.53 (m, 2 H), 7.35 (d, 7=8.70 Hz, 1 H), 8.06 (s, 1 H); m/z (ES+APCI)+: 188 [M+H]+.
Intermediate 75 l-Cyclopropyl-lH-indazol-ό-ylamine
Figure imgf000114_0003
Step l l-Cyclopropyl-ό-nitro-lH-indazole
Figure imgf000115_0001
A suspension of copper (II) acetate (1.12 g, 6.13 mmol) and [2,2']bipyridinyl (957 mg, 6.13 mmol) in hot DCE (40 ml) was added to a suspension of cyclopropylboronic acid (1.06 g, 12.27 mmol), 6- nitroindazole (1.0 g, 6.13 mmol) and sodium carbonate (1.30 g, 12.27 mmol) in DCE (20 ml). The mixture was heated at 700C for 3 hours. On cooling to it saturated ammonium chloride solution (aq) was added followed by the addition of DCM and water. The organic phase was separated and the aqueous phase re-extracted with DCM. The combined organic phases were dried and concentrated. The crude product was purified by flash column chromatography on silica gel (75 g) eluting with 3:1 petroleum etheπEtOAc to give the product as a yellow solid (626 mg, 50%). 1H NMR (400 MHz, DMSO-c?6) δ ppm 1.12 - 1.27 (m, 4 H), 3.95 - 4.02 (m, 1 H), 7.95 - 8.04 (m, 2 H), 8.27 (s, 1 H), 8.63 (s, 1 H).
Step 2 l-Cyclopropyl-lH-indazol-ό-ylamine
Figure imgf000115_0002
Prepared analogously to Intermediate 70 (Step 2) using l-cyclopropyi-6-nitro-lH-indazole to provide the product as a brown solid (542 mg, 102%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.95 - 1.08 (m, 4 H), 3.45 (tt, /=6.98, 3.55 Hz, 1 H), 5.39 (s, 2 H), 6.49 (dd, /=8.24, 1.83 Hz, 1 H), 6.59 (s, 1 H), 7.33 (d, /=9.16 Hz, 1 H), 7.64 (s, 1 H); m/z (ES+APCI)+: 174 [M+H]+.
Intermediate 76 ό-Cyclopropyl-pyridinS-ylamine
Figure imgf000115_0003
Step l
2-Cyclopropyl-5-nitro-pyridine
Figure imgf000116_0001
A mixture of 2-chloro-5-nitopyridine (1.0 g, 6.29 mmol), cyclopropylboronic acid (594 mg, 8.36 mmol), palladium (II) acetate (70 mg, 0.314 mmol), tricyclohexylphosphine (176 mg, 0.629 mmol) and potassium phosphate tribasic (4.93 g, 23.3 mmol) in water (0.3 ml) and toluene (6 ml) was placed in a RBF. The mixture was degassed, placed under an atmosphere of nitrogen and subsequently heated at 1000C overnight. On cooling to rt the mixture was diluted with EtOAc and water and was then filtered through Celite. The organic phase was washed with brine, dried and concentrated. The crude product was purified by flash column chromatography on silica gel (25 g) in 10:1 petroleum etheπEtOAc to give the product as an off-white solid (305 mg, 30%). 1H NMR (400 MHz, DMSO-40 δ ppm 1.02 - 1.18 (m, 4 H), 2.30 - 2.38 (m, 1 H), 7.62 (d, /=8.70 Hz, 1 H), 8.44 (dd, /=8.70, 2.75 Hz, 1 H), 9.20 (d, /=2.75 Hz, 1 H).
Step 2 ό-Cyclopropyl-pyridinS-ylamine
Figure imgf000116_0002
A mixture of 2-cyclopropyl-5-nitro-pyridine (300 mg, 2.24 mmol) and 10% palladium on charcoal (32 mg) in ethanol (15 ml) was stirred under a balloon of hydrogen at rt overnight. The mixture was then filtered through Celite and the filtrate was concentrated. The residue was then treated to identical hydrogenation conditions for a second time and the crude product isolated as previously. The crude material was purified by preparative HPLC (high pH buffer) to provide the product as an orange oil (30 mg, 12%). 1H NMR (400 MHz, CHLOROFORM-rf) δ ppm 0.85 - 1.06 (m, 4 H), 1.98 - 2.09 (m, 1 H), 3.78 - 4.07 (m, 2 H), 6.93 (d, /=7.79 Hz, 1 H), 7.01 (dd, /=8.24, 2.75 Hz, 1 H), 8.06 (d, /=3.21 Hz, 1 H); m/z (ES+APCI)+: 135 [M+H]+.
Intermediate 77 6-(4-Cyclopropyl-piperazin-l-yl)-pyridin-3-ylamine
Figure imgf000117_0001
l-Cyclopropyl-4-(5-nitro-pyridin-2'yl)-piperazine
Figure imgf000117_0002
A solution of DIPEA (690 μl, 3.97 mmol) and 1-cyclopropylpiperazine (500 mg, 3.97 mmol) in acetonitrile (5 ml) was added to a solution of 2-chloro-5-nitropyridine (631 mg, 3.97 mmol) in acetonitrile (10 ml). The mixture was stirred at rt for 4 hours and then concentrated to dryness. The residue was diluted with DCM and 2M sodium carbonate solution (aq). The organic phase was dried and concentrated to give a yellow solid (912 mg, 93%). 1H NMR (400 MHz, DMSO-cfe) δ ppm 0.31 - 0.49 (m, 4 H), 1.65 (it, ./=6.70, 3.38 Hz, 1 H), 2.52 - 2.69 (m, 4 H), 3.61 - 3.82 (m, 4 H), 6.95 (d, 7=9.62 Hz, 1 H), 8.21 (dd, 7=9.62, 2.75 Hz, 1 H), 8.95 (d, 7=2.75 Hz, 1 H); m/z (ES+APCI)+: 249 [M+H]+.
Step 2 6-(4-Cyclopropyl-piperazin-l-yl)-pyridin-3-ylamine
Prepared analogously Intermediate 70 (Step 2) using l-cyclopropyl-4-(5-nitro-pyridin-2-yl)- piperazine to provide the product as a dark purple coloured oil (678 mg, 85%). 1H NMR (400 MHz, DMSO-^e) δ ppm 0.27 - 0.48 (m, 4 H), 1.61 (tt, J=6.75, 3.55 Hz, 1 H), 2.54 - 2.69 (m, 4 H), 3.10 - 3.23 (m, 4 H), 4.55 (s, 2 H), 6.60 (d, 7=8.70 Hz, 1 H), 6.89 (dd, 7=8.70, 2.75 Hz, 1 H), 7.58 (d, 7=2.29 Hz, 1 H); m/z (ES+APCI)+: 219 [M+H]+. Intermediate 78
6-[2-(4-Methyl-piperazin-l-yl)-ethoxy]-pyridin-3-ylamine
Figure imgf000118_0001
Step l l-Methyl-4-[2-(5-nitro-pyridin-2-yloxy)-ethyl]-piperazine
Figure imgf000118_0002
Sodium hydride, 60% dispersion in mineral oil (252 mg, 6.29 mmol) was added, portionwise, to an ice-cooled solution of 2-(4-methyl-piperazin-l-yl)-ethanol (863 mg, 5.99 mmol) and 2-chloro-5- nitropyridine (1.0 g, 6.29 mmol) in DMF (20 ml). The ice-cooling was removed after 1 hour and the mixture was allowed to stir at rt overnight. The reaction mixture was added to ice/water and subsequently extracted with EtOAc (x2). The combined organic extracts were washed with water (x4) and brine (xl), dried and concentrated. The crude product was purified by flash column chromatography on silica gel (60 g) eluting with 10:1 DCM:MeOH to give the product as a brown oil (400 mg, 24%). 1H NMR (400 MHz, DMSO-^6) δ ppm 2.13 (s, 3 H), 2.21 - 2.54 (m, 8 H), 2.70 (t, /=5.95 Hz, 2 H), 4.50 (t, J=5.95 Hz, 2 H), 7.04 (d, .7=9.16 Hz, 1 H), 8.47 (dd, 7=9.16, 2.75 Hz, 1 H), 9.07 (d, 7=2.29 Hz, 1 H); m/z (ES+APCI)+: 267 [M+H]+.
Step 2 6-[2-(4-Methyl-piperazin-l-yl)-ethoxy]-pyridin-3-ylamine
Prepared analogously to Intermediate 70 (Step 2) using l-methyl-4-[2-(5-nitro-pyridin-2-yloxy)- ethyl]-piperazine to provide the product as a brown solid (396 mg, 114%). 1H NMR (400 MHz, DMS(W6) δ ppm 2.13 (s, 3 H), 2.21 - 2.51 (m, 8 H), 2.58 (t, 7=5.95 Hz, 2 H), 4.17 (t, 7=5.95 Hz, 2 H), 4.73 (br. s, 2 H), 6.52 (d, 7=8.70 Hz, 1 H), 6.98 (dd, 7=8.70, 2.75 Hz, 1 H), 7.47 (d, 7=3.66 Hz, 1 H); m/z (ES+APCI)+: 237 [M+H]+.
Intermediate 79
3-Methyl-3H-imidazo[4,5-b]pyridin-6-ylamine
Figure imgf000119_0001
Step l Benzhydrylidene-(3-methyl-3H-imidazo[4,5-b]pyridin-6-yl)-amine
Figure imgf000119_0002
A mixture of 6-bromo-3-methyl-3H-imidazo[4,5-b]pyridine (786 mg, 3.71 mmol), benzophenone imine (805 mg, 4.45 mmol), sodium ter/-butoxide (498 mg, 5.19 mmol), BINAP (17 mg, 0.028 mmol) and Pd2(dba)3 (8.5 mg, 0.0092 mmol) in toluene (15 ml) was place in a RBF, degassed and placed under an atmosphere of nitrogen. The mixture was stirred and heated at 800C overnight. On cooling to rt the mixture was diluted with EtOAc and water. The organic phase was washed with brine, dried and concentrated. The crude product was purified by flash column chromatography on silica gel (100 g) eluting with 40:1 DCM:MeOH to give the product as a yellow solid (663 mg, 57%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.74 (s, 3 H), 7.16 - 7.24 (m, 2 H), 7.26 - 7.42 (m, 4 H), 7.42 - 7.59 (m, 3 H), 7.69 (d, ./=7.33 Hz, 2 H), 7.83 (d, 7=2.29 Hz, 1 H), 8.28 (s, 1 H).
Step 2 3-Methyl-3H-imidazo[4,5-b]pyridin-6-ylamine
Figure imgf000119_0003
Benzhydrylidene-(3-methyl-3H-imidazo[4,5-b]pyridin-6-yl)-amine (735 mg, 2.36 mmol) in 2M HCl (0.4 ml) and THF (8 ml) was stirred at rt overnight. After stirring overnight more 2M HCl (0.4 ml) was added and stirring was continued at rt for 3 hours. The mixture was diluted with DCM and saturated sodium carbonate solution (aq). The organic phase was dried and concentrated. The crude product was purified by flash column chromatography on silica gel (30 g) eluting with 10:1 DCMMeOH to give the product as a white solid (65 mg, 19%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.73 (s, 3 H), 4.98 (s, 2 H), 7.16 (d, /=2.29 Hz, 1 H), 7.83 (d, /=2.75 Hz, 1 H), 8.13 (s, 1 H); m/z (ES+APCI)+: 149 [M+H]+.
Intermediate 80
1 -Methyl- lH-pyrazolo[ 3, 4-b]pyridin-5-ylamine
Figure imgf000120_0001
Step l Benzhydrylidene-(l-methyl-lH-pyrazolo[3, 4-b]pyridin-5-yl) -amine
Figure imgf000120_0002
Prepared analogously to Intermediate 79 (Step 1) using 5-bromo-l-methyl-lH-pyrazolo[3,4- b]pyridine and benzophenone imine to provide the product as a yellow solid (916 mg, 92%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.96 (s, 3 H), 7.20 (dd, /=7.56, 2.06 Hz, 2 H), 7.25 - 7.38 (m, 3 H), 7.39 - 7.59 (m, 4 H), 7.70 (d, /=6.87 Hz, 2 H), 7.96 (s, 1 H), 8.03 (d, /=2.29 Hz, 1 H); m/z (ES+APCI)+: 313 [M+H]+.
Step 2 l-Methyl-lH-pyrazolo[3,4-b]pyridin-5-ylamine
Prepared analogously to Intermediate 79 (Step 2) using benzhydrylidene-(l-methyl-lH- pyrazolo[3,4-b]pyridin-5-yl)-amine to provide the product as an off-white solid (260 mg, 60%). 1H NMR (400 MHz, DMSO-^6) δ ppm 3.95 (s, 3 H), 5.09 (s, 2 H), 7.17 (d, /=2.75 Hz, 1 H), 7.79 (s, 1 H), 8.08 (d, /=2.75 Hz, 1 H); m/z (ES+APCI)+: 149 [M+H]+.
Intermediate 81 -Cyclopropylmethyl-lH-pyrazol-^ylamine
Figure imgf000121_0001
Step l l-Cyclopropylmethyl^-nitro-lH-pyrazole
Figure imgf000121_0002
A mixture of 4-nitro-lH-pyrazole (1.0 g, 8.85 mmol), cyclopropylmethyl bromide (859 μl, 8.85 mmol) and potassium carbonate (1.22 g, 8.85 mmol) in DMF (25 ml) was stirred at 7O0C for 3 hours. On cooling to rt the mixture was diluted with EtOAc and washed with water (x4) and brine (xl), dried and concentrated. The residue was dissolved in EtOAc and washed with water (x3) and brine (xl), dried and concentrated to provide the product as a yellow oil (1.33 g, 90%). 1H NMR (400 MHz, DMSO-αy δ ppm 0.35 - 0.48 (m, 2 H), 0.48 - 0.62 (m, 2 H), 1.22 - 1.34 (m, 1 H), 4.03 (d, 7=7.33 Hz, 2 H), 8.27 (s, 1 H), 8.91 (s, 1 H).
Step 2 1 -Cyclopropylmethyl- lH-pyrazol-4-ylamine
Prepared analogously to to Intermediate 70 (Step 2) using l-cyclopropylmethyl-4-nitro-lH- pyrazole to provide the product as deep purple coloured oil (1.06 g, 97%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.23 - 0.34 (m, 2 H), 0.43 - 0.51 (m, 2 H), 1.03 - 1.17 (m, 1 H), 3.69 - 3.90 (m, 4 H), 6.87 (s, 1 H), 7.05 (s, 1 H); m/z (ES+APCI)+: 138 [M+H]+.
Example 1 N-{3-[5-Cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylamino]-propyl}-acetamide trifluoroacetic acid salt.
Figure imgf000122_0001
To a solution of intermediate 3 (35 mg, 0.10 mmol) in DMF (1 ml) was added glacial acetic acid (9 μl, 0.16 mmol), HATU (63.1 mg, 0.17 mmol) followed by NN-diisopropylethylamine (108 μl, 0.62 mmol), and the resulting solution was left to stir at room temperature overnight. The volatiles were removed under reduced pressure and the crude product was re-dissolved in 1:9 methanol - DCM (1 ml) and eluted though an Isolute-ΝH2 cartridge. The solvents were removed and the crude product purified by preparative LCMS (low pH buffer). 1H NMR (400 MHz, DMSOd6) δ ppm 0.54 - 0.60 (m, 2 H), 0.86 - 0.94 (m, 2 H), 1.48 - 1.57 (m, 1 H), 1.69 - 1.77 (m, 2 H), 1.78 (s, 3 H), 3.06 - 3.15 (m, 2 H), 3.45 - 3.53 (m, 2 H), 6.95 (td, /=8.4, 2.1 Hz, 1 H), 7.29 - 7.35 (m, 1 H), 7.37 - 7.45 (m, 1 H), 7.56 - 7.65 (m, 2 H), 7.93 (t, J=5.7 Hz, 1 H), 8.46 (br. s, 1 H), 10.25 (br. s, 1 H); m/z (ES+APCI)+: 344 [M+H]+.
Examples 2-10
Examples 2-10 were prepared analogously to Example 1 (the general structure is shown below followed by the tabulated examples).
Figure imgf000122_0002
Figure imgf000123_0001
Figure imgf000124_0002
*ΗPLC column: 21.2xl00mm (5μm) C-18 Phenomenex Gemini; flow rate: 20ml/min; run time: 10 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: 0.1 % trifluoroacetic acid is added to the water.
Example 11
Thiazole-5-carboxylic acid {3-[5-cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylamino]- propylj-amide
Figure imgf000124_0001
To a suspension of Intermediate 3 (35 mg, 0.10 mmol) in DMF (1 ml), was added thiazoIe-5- carboxylic acid (20.1 mg, 0.16 mmol), HATU (63.1 mg, 0.17 mmol) and N, N- diisopropylethylamine (108 μl, 0.62 mmol), and the resulting solution was stirred overnight. The volatiles were removed under reduced pressure and the crude product was re-dissolved in 1 :9 methanol : DCM (1 ml) and eluted through an Isolute-NH2 cartridge. After solvent removal, the crude product was purified by preparative LCMS (low pH buffer). After solvent removal, the product was re-dissolved in methanol/DCM (1:9) and eluted through an Isolute-NH2 cartridge and solvents evaporated to give a solid (19 mg, 44%). 1H NMR (400 MHz, CDCl3) δ ppm 0.41 - 0.60 (m, 2 H), 0.90 - 0.96 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.92 - 2.02 (m, 2 H), 3.51 - 3.72 (m, 2 H), 3.51 - 3.72 (m, 2 H), 5.87 (t, J=6.2 Hz, 1 H), 6.43 (t, ./=5.7 Hz, 1 H), 6.64 (td, ./=8.1, 2.1 Hz, 1 H), 6.92 (br. s, 1 H), 7.03 (d, /=6.9 Hz, 1 H), 7.16 - 7.24 (m, 1 H), 7.77 (s, 1 H), 7.82 (dt, J=12.4, 2.3 Hz, 1
H), 8.19 (s, 1 H), 8.88 (s, 1 H); m/z (ES+APCI)+: 413 [M+H]+.
Examples 12-25 Examples 12-25 were prepared analogously to example 11 (the general structure is shown below followed by the tabulated examples).
Figure imgf000125_0001
Figure imgf000125_0002
Figure imgf000126_0001
Figure imgf000127_0002
*HPLC column: 21.2xl00mm (5μm) C-18 Phenomenex Gemini; flow rate: 20ml/min; run time: 10 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: 0.1% trifluoroacetic acid is added to the water. Example 26
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylamino]- propyl) -amide
Figure imgf000127_0001
To a suspension of Intermediate 6 (110 mg, 0.326 mmol) and 3-fluoroaniline (0.425 mmol) in acetonitrile (1.27 ml) and water (0.128 ml) at room temperature was added, dropwise, 4M hydrogen chloride in dioxane (119 ml, 0.474 mmol). The reaction was stirred at 50 0C for 18 h and then quenched with water (10 ml). The pH was adjusted to 8 with saturated sodium bicarbonate solution and then extracted with DCM (3 x 60 ml). The combined organic layers were dried (MgSO4) and evaporated to dryness. The crude product was purified by preparative LCMS (high pH buffer). The product was re-purified by flash chromatography on the Biotage SP4, eluting with 0 to 3.5% methanol/DCM. This gave the desired product as a white solid (20 mg, 15%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.44 - 0.51 (m, 2 H), 0.78 - 0.86 (m, 2 H), 1.42 - 1.51 (m, 1 H), 1.80 - 1.90 (m, 2 H), 3.28 - 3.39 (m, 2 H), 3.46 - 3.54 (m, 2 H), 6.57 - 6.65 (m, 1 H), 6.89 - 6.96 (m, 1 H), 7.13 (dd, J=5.0, 3.7 Hz, 1 H), 7.16 - 7.24 (m, 1 H), 7.41 - 7.46 (m, 1 H), 7.64 (s, 1 H), 7.71 - 7.77 (m, 2 H), 7.84 - 7.91 (m, 1 H), 8.52 - 8.57 (m, 1 H), 9.16 (s, 1 H); m/z (ES+APCI)+: 412 [M+H]+.
Example 27 Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(3-trifluoromethyl-phenylamino)-pyrimidin-4- ylamino] -propyl} -amide
Figure imgf000128_0001
To a suspension of Intermediate 6 (40 mg, 0.119 mmol) and 3-trifiuoromethyl-phenylamine (19 μl, 0.154 mmol) in acetonitrile (0.46 ml) and water (0.047 ml) at room temperature was added 4M HCl in dioxane (0.043 ml, 0.172 mmol) dropwise., and the resulting mixture was stirred at 50 0C for 18 h. After evaporating to dryness the crude product was subjected to flash chromatography on a Biotage SP4 (methanol/DCM gradient) followed by preparative LCMS (high pH buffer) to give a white solid (8 mg, 15%). 1H NMR (400 MHz, DMSOd6) δ ppm 0.46 - 0.52 (m, 2 H), 0.80 - 0.87
(m, 2 H), 1.43 - 1.53 (m, 1 H), 1.79 - 1.89 (m, 2 H), 3.28 - 3.39 (m, 2 H), 3.47 - 3.56 (m, 2 H), 6.91 - 6.99 (m, 1 H), 7.11 - 7.16 (m, 2 H), 7.38 - 7.44 (m, 1 H), 7.66 (s, 1 H), 7.71 - 7.75 (m, 2 H), 7.83 - 7.87 (m, 1 H), 8.46 (s, 1 H), 8.51 - 8.58 (m, 1 H), 9.32 (br. s, 1 H); m/z (ES+APCI)+: 462
Example 28
Thiophene-2-carboxylic acid {3-[2-(4-acetylamino-phenylamino)-5-cyclopropyl-pyrimidin-4- ylamino]-propyl}-amide. Hydrochloride salt
Figure imgf000129_0001
Intermediate 6 (60 mg, 0.178 mmol), N-(4-aminophenyl)acetamide (32 mg, 0.214 mmol), glacial acetic acid (5 μl, 0.071 mmol) and n-butanol (1 ml) were combined and heated at 1000C for 18 h. The reaction mixture was filtered, and the residue washed with methanol and dried to give the title compound as a pale brown solid (19 mg, 24%). 1H ΝMR (400 MHz, DMSO-^6) δ ppm 0.43 - 0.49 (m, 2 H), 0.79 - 0.85 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.78 - 1.89 (m, 2 H), 2.00 (s, 3 H), 3.29 - 3.38 (m, 2 H), 3.45 - 3.53 (m, 2 H), 7.02 (br. s, 1 H), 7.10 - 7.15 (m, 1 H), 7.42 (d, 7=9.2 Hz, 2 H), 7.58 (s, 1 H), 7.62 (d, J=9.2 Hz, 2 H), 7.71 - 7.75 (m, 2 H), 8.50 - 8.61 (m, 1 H), 8.92 (br. s, 1 H), 9.75 (br. s, 1 H); m/z (ES+APCI)+: 451 [M+H]+.
Example 29 Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(3-hydroxy-phenylamino)-pyrimidin-4-ylamino]- propylj-amide Trifluoroacetic acid salt
Intermediate 6 (50 mg, 0.148 mmol), 3-aminophenol (19 mg, 0.178 mmol), glacial acetic acid (2 μl, 0.03 mmol) and n-butanol (1 ml) were combined and irradiated at 150 0C for 20 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was evaporated and then purified by preparative LCMS (low pH buffer) to give the desired product as the TFA salt, white solid (22 mg, 28%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.53 - 0.60 (m, 2 H), 0.89 (m, 2 H), 1.52 (m, 1 H), 1.86 (m, 2 H), 3.33 (m, 2 H), 3.39 - 3.61 (m, 2 H), 6.55 (dd, /=7.8, 1.8 Hz, 1 H), 7.01 (m, 2 H), 7.10 - 7.17 (m, 2 H), 7.55 (s, 1 H), 7.73 (m, 2 H), 8.53 - 8.62 (m, 2 H), 9.60 (br. s, 1 H), 10.11 (br. s, 1 H); m/z (ES+APCI)+: 410 [M+H]+.
Example 30
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(6-methyl-pyridin-3-ylamino)-pyrimidin-4- ylamino]-propyl}-amide
Figure imgf000131_0001
Intermediate 6 (60 mg, 0.178 mmol), 6-methyl-pyridin-3-ylamine (29 mg, 0.267 mmol), glacial acetic acid (5 μl, 0.071 mmol) and n-butanol (1 ml) were combined and heated at 100 0C for 18 h. More 6-methyl-pyridin-3-ylamine (10 mg, 0.089 mmol) was added and the reaction continued at 100 0C for 6 h. The reaction mixture was evaporated and then purified by preparative LCMS (low pH buffer), triturated with DCM, then eluted through a 1 g Isolute-NH2 cartridge with 9:1 DCM: methanol to give a white solid (12 mg, 18%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.43 - 0.49 (m, 2 H), 0.78 - 0.86 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.79 - 1.88 (m, 2 H), 2.35 (s, 3 H), 3.28 - 3.42 (m, 2 H), 3.44 - 3.52 (m, 2 H), 6.85 - 6.90 (m, 1 H), 7.05 (d, 7=8.7 Hz, 1 H), 7.14 (dd, 7=5.0, 3.7 Hz, 1 H), 7.61 (s, 1 H), 7.71 - 7.76 (m, 2 H), 8.09 (dd, J=8.5, 2.5 Hz, 1 H), 8.51 - 8.60 (m, 1 H), 8.73 (d, 7=2.7 Hz, 1 H), 8.97 (br. s, 1 H); m/z (ES+APCI)+: 409 [M+H]+.
Example 31
[3-(5-Cyclopropyl-4-{3-[(thiophene-2-carbonyl)-amino]ψropylamino}-pyrimidin-2-ylamino)- phenyl] -carbamic acid tert-butyl ester
Figure imgf000132_0001
Intermediate 6 (50 mg, 0.148 mmol), (3-aminophenyl)carbamic acid tert-buty\ ester (46 mg, 0.267 mmol), glacial acetic acid (2μl, 0.03 mmol) and n-butanol (1 ml) were combined and irradiated at 150 0C for 20 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was evaporated and subjected to preparative LCMS (low pH buffer), subjected to anion exchange chromatography using a 1 g Isolute-NH2 cartridge and then triturated with petroleum ether: diethyl ether (2:1) to give the desired product as a white solid (40 mg, 53%). 1H NMR (400 MHz, DMSO-c/6) δ ppm 0.43 - 0.49 (m, 2 H), 0.78 - 0.91 (m, 2 H), 1.41 - 1.51 (m, 10 H), 1.77 - 1.86 (m, 2 H), 3.24 - 3.36 (m, 2 H), 3.49 - 3.55 (m, 2 H), 6.77 - 6.81 (m, 1 H), 6.83 (dd, /=8.2, 0.9 Hz, 1 H), 7.00 - 7.06 (m, 1 H), 7.13 (dd, 7=5.0, 3.7 Hz, 1 H), 7.35 (dd, 7=8.0, 1.1 Hz, 1 H), 7.59 (s, 1 H), 7.69 - 7.75 (m, 2 H), 7.91 (s, 1 H), 8.50 - 8.56 (m, 1 H), 8.79 (s, 1 H), 9.15 (br. s, 1 H); m/z (ES+APCI)+: 409 [M- CO2'Bu+2H]+.
Example 32 Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(3-methanesulfonylamino-phenylamino)- pyrimidin-4-ylamino]-propyl}-amide triβuoroacetic acid salt
Figure imgf000133_0001
Intermediate 6 (41 mg, 0.121 mmol), N-(3-amino-phenyl)-methane sulfonamide (68 mg, 0.365 mmol), glacial acetic acid (8 μl, 0.121 mmol) and n-butanol (1.3 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was evaporated and then purified by preparative LCMS (low pH buffer) to give a white solid (42 mg, 57%). 1H ΝMR (400 MHz, DMSO-Cf6) δ ppm 0.53 - 0.61 (m, 2 H), 0.86 - 0.93 (m, 2 H), 1.49 - 1.57 (m, 1 H), 1.80 - 1.89 (m, 2 H), 3.00 (s, 3 H), 3.27 - 3.35 (m, 2 H), 3.53 - 3.61 (m, 2 H), 6.92 - 6.97 (m, 1 H), 7.13 (dd, 7=4.8, 3.9 Hz, 1 H), 7.27 - 7.34 (m, 1 H), 7.36 - 7.42 (m, 2 H), 7.58 (s, 1 H), 7.68 - 7.76 (m, 2 H), 8.46 - 8.60 (m, 2 H), 9.86 (s, 1 H), 10.20 (br. s, 1 H); m/z (ES+APCI)+: 487 [M+H]+.
Examples 33-36
Examples 33-36 were prepared analogously to example 32 (the general structure is shown below followed by the tabulated examples).
Figure imgf000134_0001
Figure imgf000135_0003
*HPLC column: 21.2xl00mm (5μm) C-18 Phenomenex Gemini; flow rate: 20ml/min; run time: 10 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: 0.1% trifluoroacetic acid is added to the water.
Example 37
Thiophene-2-carboxylic acid {3-[2-(benzothiazol-6-ylamino)-5-cyclopropyl-pyrimidin-4-ylamino]- propyl} -amide .Hydrochloride salt
Figure imgf000135_0001
Intermediate 6 (41 mg, 0.121 mmol), benzothiazol-6-ylamine (55 mg, 0.365 mmol), glacial acetic acid (8 μl, 0.121 mmol) and n-butanol (1.3 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was filtered, washed with methanol and the resultant solid was dried to give the desired product as a white solid (25 mg, 46%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.54 - 0.60 (m, 2 H), 0.88 - 0.95 (m, 2 H), 1.50 - 1.59 (m, 1 H), 1.82 - 1.92 (m, 2 H), 3.29 - 3.36 (m, 2 H), 3.50 - 3.58 (m, 2 H), 7.11 (dd, J=4.8, 3.9 Hz, 1 H), 7.59 (s, 1 H), 7.64 (dd, 7=8.7, 1.8 Hz, 1 H), 7.72 - 7.75 (m, 2 H), 8.08 (d, J=9.2 Hz, 1 H), 8.40 (d, 7=1.8 Hz, 1 H), 8.60 - 8.66 (m, 2 H), 9.33 (s, 1 H), 10.40 (br. s, 1 H); m/z (ES+APCI)+: 451 [M+H]+.
Example 38 l-{3-[5-Cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylamino]-propyl}-pyrrolidin-2-one
Figure imgf000135_0002
Intermediate 7 (97 mg, 0.33 mmol), 3-fluororaniline (94 μl, 0.99 mmol) and glacial acetic acid (60 μl, 1.06 mmol) were combined in n-butanol (1 ml) and heated in the microwave for 40 minutes at
140 0C. The solvents were removed under reduced pressure and the crude product was purified by preparative LCMS (low pH buffer) and the resulting product subjected to anion exchange chromatography using an Isolute-NH2 cartridge to give a white solid (46 mg, 38%). 1H NMR (400
MHz, DMSO-J6) δ ppm 0.46 - 0.51 (m, 2 H), 0.78 - 0.90 (m, 2 H), 1.44 - 1.52 (m, 1 H), 1.79 (quin,
/=6.9 Hz, 2 H), 1.85 - 1.98 (m, 2 H), 2.14 - 2.27 (m, 2 H), 3.23 - 3.31 (m, 2 H), 3.35 (t, /=7.1 Hz, 2
H), 3.43 (q, 7=6.4 Hz, 2 H), 6.62 (td, /=8.2, 2.3 Hz, 1 H), 6.84 (t, /=6.0 Hz, 1 H), 7.18 - 7.25 (m, 1
H), 7.44 (d, /=8.2 Hz, 1 H), 7.65 (s, 1 H), 7.79 - 7.86 (m, 1 H), 9.08 (s, 1 H); m/z (ES+APCI)+: 370 [M+H]+
Example 39
Η-β-{5-Cyclopropyl-4-[3-(2-oxo-pyrrolidinA-yl)-propylamino]ψyrimidin-2-ylcmino}-phenyl)- acetamide trifluoroacetate salt
Figure imgf000136_0001
Intermediate 7 (97 mg, 0.33 mmol), 3-aminoacetanilide (148 mg, 0.99 mmol) and glacial acetic acid (60 μl, 1.06 mmol) were combined in M-butanol (1 ml) and heated in the microwave for 1 hour at 150 0C. The solvents were removed under reduced pressure and the crude product was purified by preparative LCMS (low pH). The resulting TFA salt was subjected anion exchange chromatography using an Isolute-NH2 cartridge to give a white solid (46 mg, 33%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.44 - 0.50 (m, 2 H), 0.76 - 0.88 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.61 - 1.82 (m, 2 H), 1.86 - 1.95 (m, 2 H), 2.01 (s, 3 H), 2.14 - 2.28 (m, 2 H), 3.25 (t, /=6.9 Hz, 2 H), 3.30 - 3.35 (m, 2 H), 3.43 (q, /=6.4 Hz, 2 H), 6.84 (t, /=5.7 Hz, 1 H), 7.03 - 7.13 (m, 2 H), 7.39 (d, /=7.3 Hz, 1 H), 7.59 (s, 1 H), 7.92 (s, 1 H), 8.91 (s, 1 H), 9.80 (s, 1 H); m/z (ES+APCI)+: 409 [M+H]+.
Example 40 Cyclobutanecarboxylic acid {3-[2-(benzothiazol-6-ylamino)-5-cyclopropyl-pyrimidin-4-ylamino]- propyl} -amide. Hydrochloride salt.
Figure imgf000137_0001
Intermediate 8 (50 mg, 0.162 mmol), benzothiazol-6-ylamine (73 mg, 0.486 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was filtered, the residue was washed with methanol and dried to give the product as a white solid (52 mg, 76%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.54 - 0.61 (m, 2 H), 0.87 - 0.95 (m, 2 H), 1.51 - 1.60 (m, 1 H), 1.65 - 1.78 (m, 3 H), 1.78 - 1.90 (m, 1 H), 1.90 - 2.00 (m, 2 H), 2.02 - 2.15 (m, 2 H), 2.88 - 2.99 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.39 - 3.55 (m, 2 H), 7.61 - 7.70 (m, 2 H), 7.75 - 7.82 (m, 1 H), 8.11 (d, J=8.7 Hz, 1 H), 8.41 (d, /=2.3 Hz, 1 H), 8.64 - 8.73 (m, 1 H), 9.34 (s, 1 H), 10.68 (br. s, 1 H); m/z (ES+APCI)+: 423 [M+H]+.
Example 41
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-morpholin-4-yl-phenylamino)-pyrimidin-4- ylamino] -propyl} -amide
Figure imgf000137_0002
Intermediate 8 (50 mg, 0.162 mmol), 3-morpholin-4-yl-phenylamine (86 mg, 0.486 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was evaporated and subjected to preparative LCMS (low pH buffer). The resulting TFA salt was subjected anion exchange chromatography using an Isolute-NH2 cartridge to give a white solid (39 mg, 53%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.43 - 0.49 (m, 2 H), 0.77 - 0.87 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.62 - 1.78 (m, 3 H), 1.79 - 1.91 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.90 - 3.00 (m, 1 H), 3.02 - 3.07 (m, 4 H), 3.09 - 3.16 (m, 2 H), 3.40 - 3.47 (m, 2 H), 3.70 - 3.75 (m, 4 H), 6.45 (dd, 7=8.2, 1.8 Hz, 1 H), 6.77 - 6.82 (m, 1 H), 7.01 - 7.08 (m, 1 H), 7.22 (dd, /=8.0, 1.1 Hz, 1 H), 7.44 - 7.47 (m, 1 H), 7.60 (s, 1 H), 7.65 - 7.70 (m, 1 H), 8.72 (br. s, 1 H); m/z (ES+APCI)+: 451.
Example 42
Cyclobutanecarboxylic acid {3-[5~cyclopropyl-2-(3-imidazol-l-ylmethyl-phenylamino)-pyrimidin- 4-ylarnino] -propyl} -amide
Figure imgf000138_0001
Prepared analogously to example 41 from Intermediate 8 and 3-(lH-imidazol-l-ylmethyl)aniline to give a white solid (7 mg, 10%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.44 - 0.48 (m, 2 H), 0.79 - 0.85 (m, 2 H), 1.42 - 1.49 (m, 1 H), 1.64 - 1.76 (m, 3 H), 1.80 - 1.89 (m, 1 H), 1.92 - 2.01 (m, 2 H), 2.04 - 2.15 (m, 2 H), 2.90 - 3.00 (m, 1 H), 3.09 - 3.15 (m, 2 H), 3.36 - 3.42 (m, 2 H), 5.10 - 5.14 (m, 2 H), 6.64 - 6.68 (m, 1 H), 6.82 - 6.86 (m, 1 H), 6.89 - 6.90 (m, 1 H), 7.11 - 7.14 (m, 1 H), 7.15 - 7.20 (m, 1 H), 7.60 (s, 1 H), 7.63 (dd, /=8.2, 1.4 Hz, 1 H), 7.67 - 7.76 (m, 3 H), 8.96 (br. s, 1 H); m/z (ES+APCI)+: 446.
Example 43
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(4-methyl-piperazin-l-yl)-pyridin-3-ylamino]- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000139_0001
Intermediate 8 (50 mg, 0.162 mmol), 6-(4-methyl-piperazin-l-yl)-pyridin-3-ylamine (93 mg, 0.486 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 15O0C for 40 minutes in a Biotage 1-60 microwave reactor. Glacial acetic acid (27μl, 0.486 mmol) was added and the reaction was irradiated at 15O0C for a further 40 minutes. The reaction mixture was evaporated and then purified by preparative LCMS (low pH buffer), and the resulting TFA salt was subjected anion exchange chromatography using an Isolute-NH2 cartridge to give the product as a white solid (14.4 mg, 19%). 1H NMR (400 MHz, DMSO-Cf6) δ ppm 0.40 - 0.46 (m, 2 H), 0.77 - 0.84 (m, 2 H), 1.39 - 1.48 (m, 1 H), 1.60 - 1.78 (m, 3 H), 1.79 - 1.92 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.04 - 2.17 (m, 2 H), 2.20 (s, 3 H), 2.36 - 2.43 (m, 4 H), 2.46 - 2.54 (m, 4 H), 2.91 - 3.00 (m, 1 H), 3.07 - 3.16 (m, 2 H), 3.29 - 3.42 (m, 2 H), 6.73 - 6.80 (m, 2 H), 7.55 (s, 1 H), 7.65 - 7.70 (m, 1 H), 7.94 (dd, 7=9.2, 2.7 Hz, 1 H), 8.41 (d, 7=2.3 Hz, 1 H), 8.63 (br. s, 1 H); m/z (ES+APCI)+: 465.
Example 44
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(3-dimethylamino-phenylamino)-pyrimidin-4- ylamino]-propyl}-amide trifluoroacetic acid salt.
Figure imgf000140_0001
Intermediate 6 (41 mg, 0.122 mmol), N,N-dimethyl-benzene- 1,3 -diamine (61 mg, 0.365 mmol), glacial acetic acid (8μl, 0.122 mmol) and n-butanol (1.3 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was evaporated and subjected to preparative LCMS (low pH buffer) followed by flash chromatography on the Biotage SP4 (gradient elution from 0 to 8% methanol in DCM) gave a white solid (5 mg, 7%). 1H ΝMR (400 MHz, DMSO-^6) δ ppm 0.43 - 0.49 (m, 2 H), 0.78 - 0.85 (m, 2 H), 1.41 - 1.51 (m, 1 H), 1.78 - 1.87 (m, 2 H), 2.85 (s, 6 H), 3.34 (s, 2 H), 3.48 - 3.55 (m, 2 H), 6.24 (dd, 7=8.2, 2.3 Hz, 1 H), 6.74 - 6.81 (m, 1 H), 6.95 - 7.00 (m, 1 H), 7.08 - 7.11 (m, 1 H), 7.13 (dd, J=5.0, 3.7 Hz, 1 H), 7.24 - 7.27 (m, 1 H), 7.60 (s, 1 H), 7.69 - 7.76 (m, 2 H), 8.51 - 8.57 (m, 1 H), 8.63 (br. s, 1 H); m/z (ES+APCI)+: 437 [M+H]+.
Example 45
Thiophene-2-carboxylic acid (3-{5-cyclopropyl-2-[3-(4-methyl-piperazine-l-carbonyl)- phenylamino]-pyrimidin-4-ylamino} -propyl) -amide
Figure imgf000140_0002
To a solution of Example 34 (20.1 mg, 0.036 mmol) in DMF (0.5 ml) was added 1- methylpiperazine (6 μl, 0.055 mmol), HATU (22 mg, 0.58 mmol) and Ν,Ν-diisopropylethylamine (38 μl, 0.219 mmol). The reaction was stirred at room temperature for 18 hours. The mixture was evaporated then filtered through a 1 g Isolute-NH2 cartridge, eluting with 9:1 DCM : methanol. Purification by flash chromatography on the Biotage SP4 (gradient elution from 0 to 10% methanol in DCM) gave the desired product as a white solid (18.9 mg, 98%). 1H NMR (400 MHz, DMS0-<4 85 0C) δ ppm 0.45 - 0.52 (m, 2 H), 0.81 - 0.89 (m, 2 H), 1.46 - 1.55 (m, 1 H), 1.83 - 1.93 (m, 2 H), 2.28 (s, 3 H), 2.39 - 2.46 (m, 4 H), 3.33 - 3.42 (m, 2 H), 3.46 - 3.57 (m, 6 H), 6.58 - 6.65 (m, 1 H), 6.83 (ddd, 7=7.6, 1.4, 1.1 Hz, 1 H), 7.11 (dd, 7=4.8, 3.9 Hz, 1 H), 7.22 - 7.28 (m, 1 H), 7.63 (s, 1 H), 7.66 - 7.72 (m, 2 H), 7.76 (ddd, 7=8.2, 2.3, 0.9 Hz, 1 H), 7.86 - 7.90 (m, 1 H), 8.22 - 8.28 (m, 1 H), 8.73 (br. s, 1 H); m/z (ES+ APCI)+: 520 [M+H]+.
Example 46
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(3-morpholin-4-yl-phenylamino)-pyrimidin-4- ylamino]-propyl}-amide trifluoroacetic acid salt.
Figure imgf000141_0001
Prepared analogously to example 44 from Intermediate 6 and 3-morpholin-4-ylaniline. The product was isolated as a white foam (25 mg, 35%). 1H NMR (400 MHz, DMSO-cfe) δ ppm 0.44 - 0.50 (m, 2 H), 0.78 - 0.85 (m, 2 H), 1.42 - 1.51 (m, 1 H), 1.78 - 1.88 (m, 2 H), 3.01 - 3.08 (m, 4 H), 3.29 - 3.38 (m, 2 H), 3.46 - 3.55 (m, 2 H), 3.70 - 3.76 (m, 4 H), 6.44 (dd, 7=8.0, 2.1 Hz, 1 H), 6.79 - 6.83 (m, 1 H), 6.99 - 7.06 (m, 1 H), 7.13 (dd, 7=5.0, 3.7 Hz, 1 H), 7.23 (dd, 7=8.0, 1.1 Hz, 1 H), 7.43 - 7.47 (m, 1 H), 7.61 (s, 1 H), 7.71 - 7.76 (m, 2 H), 8.52 - 8.57 (m, 1 H), 8.72 (br. s, 1 H); m/z (ES+APCI)+: 479 [M+H]+.
Example 47
Thiophene-2-carboxylic acid (3-{5-cyclopropyl-2-[3-(2-oxo-pyrrolidin-l-yl)-phenylaminoJ- pyrimidin-4-ylamino}-propyl)-amide. Hydrochloride salt
Figure imgf000142_0001
Intermediate 6 (40 mg, 0.119 mmol), l-(3-amino-phenyl)-pyrrolidin-2-one (63 mg, 0.356 mmol), glacial acetic acid (8 μl, 0.119 mmol) and n-butanol (1 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was evaporated, the residue was triturated with methanol and dried to yield a white solid (12.4 mg, 22%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.52 - 0.60 (m, 2 H), 0.87 - 0.94 (m, 2 H), 1.50 - 1.60 (m, 1 H), 1.78 - 1.89 (m, 2 H), 2.00 - 2.12 (m, 2 H), 3.26 - 3.33 (m, 2 H), 3.33 - 3.46 (m, 2 H), 3.52 - 3.63 (m, 2 H), 3.78 - 3.88 (m, 2 H), 7.12 (dd, 7=4.8, 3.9 Hz, 1 H), 7.23 - 7.40 (m, 3 H), 7.60 (s, 1 H), 7.70 - 7.77 (m, 2 H), 8.04 - 8.11 (m, 1 H), 8.57 - 8.70 (m, 2 H), 10.29 (br. s, 1 H); m/z (ES+APCI)+: 477 [M+H]+.
Example 48
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[3-(2-oxo-pyrrolidin-l-yl)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000143_0001
Intermediate 8 (50 mg, 0.162 mmol), l-(3-aminophenyl)-pyrrolidin-2-one (86 mg, 0.485 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The reaction mixture was evaporated, the residue was triturated with methanol and the resultant solid was dried to yield the title compound as a white solid (27 mg, 37%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.53 - 0.60 (m, 2 H), 0.87 - 0.94 (m, 2 H), 1.50 - 1.60 (m, 1 H), 1.65 - 1.77 (m, 3 H), 1.78 - 1.91 (m, 1 H), 1.91 - 2.01 (m, 2 H), 2.02 - 2.14 (m, 4 H), 2.87 - 3.00 (m, 1 H), 3.05 - 3.13 (m, 2 H), 3.32 - 3.43 (m, 2 H), 3.47 - 3.56 (m, 2 H), 3.80 - 3.87 (m, 2 H), 7.25 - 7.43 (m, 3 H), 7.63 (s, 1 H), 7.73 - 7.81 (m, 1 H), 8.06 - 8.12 (m, 1 H), 8.56 - 8.67 (m, 1 H), 10.42 (br. s, 1 H); m/z (ES+APCI)+: 449 [M+H]+.
Example 49
Pyrrolidine- 1-carboxylic acid [3-(5-cyclopropyl-4-{3-[(thiophene-2-carbonyl)-amino]- propylamino}-pyrimidin-2-ylamino)-phenyl] -amide
Figure imgf000144_0001
Intermediate 6 (50 mg, 0.148 mmol), Intermediate 10 (43 mg, 0.178 mmol), tris(dibenzylideneacetone)dipalladium(0) (8 mg, 0.009 rnmol), xantphos (7 mg, 0.012 mmol) and sodium tert-butoxide (43 mg, 0.44 mmol) were combined with dioxane (3 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 105 0C for 18 h. The reaction mixture was evaporated onto silica, then subjected to flash chromatography on the Biotage SP4 (gradient elution from 0 to 10% methanol in DCM). Further purification by cation exchange chromatography using an Isolute SCX cartridge gave a white solid (9 mg, 12%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.42 - 0.48 (m, 2 H), 0.78 - 0.85 (m, 2 H), 1.40 - 1.50 (m, 1 H), 1.77 - 1.87 (m, 6 H), 3.24 - 3.61 (m, 8 H), 6.76 - 6.81 (m, 1 H), 6.88 - 6.92 (m, 1 H), 6.99 - 7.04 (m, 1 H), 7.10 - 7.14 (m, 1 H), 7.27 - 7.32 (m, 1 H), 7.59 (s, 1 H), 7.69 - 7.75 (m, 2 H), 7.86 - 7.90 (m, 1 H), 7.94 (s, 1 H), 8.52 - 8.56 (m, 1 H), 8.75 (br. s, 1 H); m/z (ES+APCI)+: 506 [M+H]+.
Example 50
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(pyridin-3-ylamino)-pyrimidin-4-ylamino]- propyl} -amide
Figure imgf000145_0001
Intermediate 6 (60 mg, 0.179 mmol), 3-aminopyridine (20 mg, 0.213 mmol), tris- (dibenzylideneacetone)dipalladium(O) (9.8 mg, 0.011 mmol), xantphos (8 mg, 0.014 mmol) and sodium tert-butoxide (48 mg, 0.5 mmol) were combined with dioxane (3 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 105 0C for 18 h. The reaction mixture was evaporated onto silica, then purified by flash chromatography on a Biotage SP4 (gradient elution from 0 to 10% methanol in DCM) to give an off-white solid (18 mg, 26%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.44 - 0.51 (m, 2 H), 0.83 (m, 2 H), 1.47 (m, 1 H), 1.79 - 1.89 (m, 2 H), 3.34 (m, 2 H), 3.49 (m, 2 H), 6.92 (m, 1 H), 7.13 (dd, J=5.0, 3.7 Hz, 1 H), 7.21 (dd, J=8.5, 4.8 Hz, 1 H), 7.64 (s, 1 H), 7.70 - 7.76 (m, 2 H), 8.03 (dd, ./=4.6, 1.4 Hz, 1 H), 8.21 - 8.26 (m, 1 H), 8.53 - 8.59 (m, 1 H), 8.84 - 8.87 (m, 1 H), 9.10 (s, 1 H); m/z (ES+APCI)+: 395 [M+H]+.
Example 51 Thiophene-2-carboxylic acid {3-[2-(3-acetylamino-phenylamino)-5-cyclopropyl-pyrimidin-4- ylamino]-propyl}-amide
Figure imgf000146_0001
Intermediate 6 (60 mg, 0.179 mmol), N-(3-aminophenyl)acetamide (32 mg, 0.215 mmol), tris(dibenzylideneacetone)dipalladium(0) (9.8 mg, 0.011 mmol), xantphos (8 mg, 0.014 mmol) and sodium tert-butoxide (48 mg, 0.5 mmol) were suspended in dioxane (3 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 105 0C for 18 h. The reaction mixture was evaporated onto silica subjected to flash chromatography on the Biotage SP4 (gradient elution from 0 to 10% methanol in DCM). Further purification by preparative LCMS (high pH buffer) gave the desired product as a white solid (7 mg, 9%). 1H ΝMR (400 MHz, DMSO-J6) δ ppm 0.43 - 0.49 (m, 2 H), 0.78 - 0.85 (m, 2 H), 1.42 - 1.51 (m, 1 H), 1.78 - 1.87 (m, 2 H), 2.01 (s, 3 H), 3.29 - 3.37 (m, 2 H), 3.46 - 3.56 (m, 2 H), 6.76 - 6.82 (m, 1 H), 7.02 - 7.11 (m, 2 H), 7.13 (dd, /=4.8, 3.9 Hz, 1 H), 7.39 - 7.43 (m, 1 H), 7.60 (s, 1 H), 7.70 - 7.76 (m, 2 H), 7.93 - 7.95 (m, 1 H), 8.51 - 8.57 (m, 1 H), 8.85 (br. s, 1 H), 9.77 (br. s, 1 H); m/z (ES+APCI)+: 451 [M+H]+.
Example 52 Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(2-fluoro-phenylamino)-pyrimidin-4-ylamino]- propyl}-amide
Figure imgf000147_0001
Intermediate 6 (60 mg, 0.179 mmol), 2-fluorophenylamine (21 mg, 0.215 mmol), palladium (II) acetate (1.6 mg, 0.007 mmol), xantphos (8 mg, 0.014 mmol) and cesium carbonate (174 mg, 0.5 mmol) were combined with dioxane (3 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 105 0C for 18 h. The reaction mixture was evaporated onto silica then subjected to flash chromatography on the Biotage SP4 (gradient elution from 0 to 10% methanol in DCM). Further purification by preparative LCMS (high pH buffer) gave a white solid (4 mg, 5%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.41 - 0.50 (m, 2 H), 0.77 - 0.85 (m, 2 H), 1.40 - 1.49 (m, 1 H), 1.73 - 1.85 (m, 2 H), 3.22 - 3.39 (m, 2 H), 3.39 - 3.48 (m, 2 H), 6.82 - 6.90 (m, 1 H), 6.91 - 6.98 (m, 1 H), 7.05 - 7.10 (m, 1 H), 7.11 - 7.19 (m, 2 H), 7.58 (s, 1 H), 7.69 - 7.77 (m, 2 H), 8.02 (br. s, 1 H), 8.04 - 8.10 (m, 1 H), 8.45 - 8.60 (m, 1 H); m/z (ES+APCI)+: 412 [M+H]+.
Example 53
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(isoquinolin-6-ylamino)-pyrimidin-4-ylamino]- propylj-amide
Figure imgf000147_0002
Intermediate 6 (50 mg, 0.148 mmol), Isoquinolin-6-ylamine (26 mg, 0.178 mmol), tris(dibenzylideneacetone)dipalladium(0) (8 mg, 0.009 mmol), xantphos (7 mg, 0.012 mmol) and sodium tert-butoxide (43 mg, 0.44 mmol) were combined with dioxane (3 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 1050C for 18 h, evaporated and purified through a silica plug, eluting with 0 to 10% methanol/DCM. Further purification by preparative LCMS (high pH buffer) gave the desired product as a white solid (15 mg, 23%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.42 - 0.50 (m, 2 H), 0.78 - 0.86 (m, 2 H), 1.41 - 1.52 (m, 1 H), 1.71 - 1.81 (m, 2 H), 3.24 - 3.31 (m, 2 H), 3.36 - 3.45 (m, 2 H), 6.81 - 6.89 (m, 1 H), 7.13 (dd, 7=4.8, 3.9 Hz, 1 H), 7.54 - 7.63 (m, 2 H), 7.70 - 7.76 (m, 3 H), 8.02 (d, /=6.4 Hz, 1 H), 8.24 (dd, /=7.8, 0.9 Hz, 1 H), 8.43 (d, /=6.0 Hz, 1 H), 8.48 - 8.55 (m, 1 H), 8.90 (s, 1 H), 9.23 (s, 1 H); m/z (ES+APCI)+: 445 [M+H]+.
Examples 54-55
Examples 54-55 were prepared analogously to Example 53 (the general structure is shown below followed by the tabulated examples).
Figure imgf000149_0002
*HPLC column: 21.2xl00mm (lOμm) C-18 Phenomenex Gemini; flow rate: 20ml/min; run time: 10 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: ammonium bicarbonate (10 mmol) and ammonium hydroxide is added to the water.
Example 56
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(quinoxalin-6-ylamino)-pyrimidin-4-ylamino]- propylj-amide
Figure imgf000149_0001
Prepared analogously to Example 50 from Intermediate 6 and quinoxalin-6-ylamine to give desired product as a yellow solid (25 mg, 38%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.49 - 0.55 (m, 2 H), 0.82 - 0.89 (m, 2 H), 1.46 - 1.56 (m, 1 H), 1.87 - 1.97 (m, 2 H), 3.36 - 3.45 (m, 2 H), 3.55 - 3.64 (m, 2 H), 6.99 - 7.05 (m, 1 H), 7.11 (dd, 7=4.8, 3.9 Hz, 1 H), 7.69 - 7.75 (m, 3 H), 7.91 (d, ./=9.2 Hz, 1 H), 8.04 (dd, J=9.2, 2.3 Hz, 1 H), 8.52 - 8.58 (m, 1 H), 8.66 (d, /=1.8 Hz, 1 H), 8.77 (d, J=I.8 Hz, 1 H), 8.82 (d, 7=2.3 Hz, 1 H), 9.58 (br. s, 1 H); m/z (ES+APCI)+: 446 [M+H]+.
Example 57
Thiophene-2-carboxylic acid [3-(5-cyclopropyl-2-phenylamino-pyrimidin-4-ylamino)-propyl]- amide
Figure imgf000150_0001
Prepared analogously to Example 50 from Intermediate 6 and phenylamine to give the desired product as a white solid (20 mg, 9%). 1H NMR (400 MHz, DMSO-rf6) δ ppm 0.41 - 0.51 (m, 2 H), 0.77 - 0.87 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.78 - 1.89 (m, 2 H), 3.23 - 3.41 (m, 2 H), 3.43 - 3.53 (m, 2 H), 6.77 - 6.86 (m, 2 H), 7.13 (dd, 7=5.0, 3.7 Hz, 1 H), 7.15 - 7.21 (m, 2 H), 7.61 (s, 1 H), 7.68 - 7.78 (m, 4 H), 8.50 - 8.57 (m, 1 H), 8.87 (br. s, 1 H); m/z (ES+APCI)+: 394 [M+H]+.
Example 58 Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(4-fluoro-phenylamino)-pyrimidin-4-ylamino]- propyl}-amide
Figure imgf000150_0002
Prepared analogously to Example 50 from Intermediate 6 and 4-fluorophenylamine to give desired product as an off-white solid (25 mg, 41%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.36 - 0.53 (m, 2 H), 0.67 - 0.91 (m, 2 H), 1.38 - 1.51 (m, 1 H), 1.67 - 1.91 (m, 2 H), 3.19 - 3.43 (m, 2 H), 3.43 - 3.61 (m, 2 H), 6.73 - 6.92 (m, 1 H), 6.92 - 7.08 (m, 2 H), 7.13 (dd, 7=4.8, 3.9 Hz, 1 H), 7.60 (s, 1 H), 7.67 - 7.76 (m, 4 H), 8.43 - 8.61 (m, 1 H), 8.85 - 8.99 (m, 1 H); m/z (ES+APCI)+: 412 [M+H]+.
Example 59
Thiophene-2-carboxylic acid [3-(5-cyclopropyl-2-o-tolylamino-pyrimidin-4-ylamino)-propyl]- amide
Figure imgf000151_0001
Prepared analogously to Example 50 from Intermediate 6 and ø-tolylamine to give desired product as an off-white solid (10 mg, 17%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.41 - 0.46 (m, 2 H), 0.77 - 0.83 (m, 2 H), 1.40 - 1.47 (m, 1 H), 1.73 - 1.81 (m, 2 H), 2.21 (s, 3 H), 3.26 - 3.36 (m, 2 H), 3.37 - 3.45 (m, 2 H), 6.71 - 6.76 (m, 1 H), 6.86 - 6.91 (m, 1 H), 7.04 - 7.16 (m, 3 H), 7.54 (s, 1 H), 7.69 - 7.76 (m, 4 H), 8.46 - 8.58 (m, 1 H); m/z (ES+APCI)+: 408 [M+H]+.
Example 60
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(2-methyl-benzothiazol-5-ylamino)-pyrimidin-4- ylamino]-propyl}-amide
Figure imgf000152_0001
Prepared analogously to Example 50 from Intermediate 6 and 2-Methyl-benzothiazol-5-ylamine gave the desired product as a white solid (7 mg, 10%). 1H NMR (400 MHz, DMSO-^6, 850C) δ ppm 0.47 - 0.51 (m, 2 H), 0.82 - 0.88 (m, 2 H), 1.48 - 1.55 (m, 1 H), 1.88 - 1.96 (m, 2 H), 2.74 (s, 3 H), 3.38 - 3.44 (m, 2 H), 3.54 - 3.60 (m, 2 H), 6.56 - 6.60 (m, 1 H), 7.10 (dd, J=5.0, 3.7 Hz, 1 H), 7.65 (s, 1 H), 7.66 - 7.70 (m, 2 H), 7.72 - 7.74 (m, 2 H), 8.25 - 8.29 (m, 1 H), 8.46 - 8.48 (m, 1 H), 8.73 (br. s, 1 H); m/z (ES+APCI)+: 465 [M+H]+.
Example 61
Thiophene-2-carboxylic acid β-ft-β-cyano-phenylaminoJ-S-cyclopropyl-pyrimidin^-ylaminoJ- propyl} -amide
Figure imgf000152_0002
Prepared analogously to Example 50 from Intermediate 6 and 3-amino benzonitrile to give desired product as a white solid (15 mg, 24%). 1H NMR (400 MHz, DMS(W6) δ ppm 0.46 - 0.51 (m, 2 H), 0.80 - 0.87 (m, 2 H), 1.44 - 1.51 (m, 1 H), 1.81 - 1.89 (m, 2 H), 3.26 - 3.41 (m, 2 H), 3.45 - 3.59 (m, 2 H), 6.95 - 7.00 (m, 1 H), 7.13 (dd, 7=4.8, 3.9 Hz, 1 H), 7.23 - 7.28 (m, 1 H), 7.38 - 7.43 (m, 1 H), 7.67 (s, 1 H), 7.71 - 7.76 (m, 2 H), 7.92 - 7.96 (m, 1 H), 8.38 - 8.41 (m, 1 H), 8.52 - 8.57 (m, 1 H),
9.33 (br. s, 1 H); m/z (ES+APCI)+: 419 [M+H]+.
Example 62 Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(2-trifluoromethyl-lH-benzimidazol-5-ylamino)- pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000153_0001
Prepared analogously to Example 28 from Intermediate 6 and 2-trifluoromethyl-lH-benzimidazol- 5-ylamine to give the desired product as a white solid (10 mg, 13%). 1H NMR (400 MHz, DMSO- d6) δ ppm 0.46 - 0.52 (m, 2 H), 0.80 - 0.87 (m, 2 H), 1.45 - 1.52 (m, 1 H), 1.84 - 1.92 (m, 2 H), 3.21 - 3.45 (m, 2 H), 3.46 - 3.58 (m, 2 H), 6.84 - 6.89 (m, 1 H), 7.12 (dd, 7=5.0, 3.7 Hz, 1 H), 7.42 - 7.63 (m, 2 H), 7.65 (s, 1 H), 7.68 - 7.75 (m, 2 H), 8.43 - 8.47 (m, 1 H), 8.49 - 8.64 (m, 2 H), 9.13 (br. s, 1 H); m/z (ES+APCI)+: 502 [M+H]+.
Example 63
Thiophene-2-carboxylic acid {3-[5-cyclopropyl-2-(pyridin-4-ylamino)-pyrimidin-4-ylamino]- propylj-amide
Figure imgf000154_0001
Prepared analogously to Example 51 from Intermediate 6 and 4-aminopyridine. m/z (ES+ APCl/ 395 [M+H]\
Example 64
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(5-fluoro-pyridin-3-ylamino)-pyrimidin-4- ylaminoj-propylj-amide
Figure imgf000154_0002
Intermediate 8 (45 mg, 0.146 mmol), 3-amino-5-fluoropyridine (20 mg, 0.175 mmol), tris(dibenzylideneacetone)dipalladium(0) (8 mg, 0.009 mmol), xantphos (7 mg, 0.012 mmol) and sodium tert-butoxide (42 mg, 0.437 mmol) were combined with dioxane (3 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 100 0C for 18 hours, evaporated and purified through a silica plug, eluting with 0 to 10% methanol/DCM. Further purification by preparative LCMS (low pH buffer) followed by anion exchange chromatography of the resulting TFA salt using an Isolute-NH2 cartridge to give the product as a white solid (28 mg, 50%). 1H
NMR (400 MHz, DMSO--4) δ ppm 0.46 - 0.51 (m, 2 H), 0.81 - 0.88 (m, 2 H), 1.40 - 1.53 (m, 1 H),
1.64 - 1.78 (m, 3 H), 1.79 - 1.92 (m, 1 H), 1.92 - 2.03 (m, 2 H), 2.03 - 2.18 (m, 2 H), 2.91 - 3.02 (m,
1 H), 3.08 - 3.20 (m, 2 H), 3.38 - 3.47 (m, 2 H), 6.97 - 7.07 (m, 1 H), 7.67 (s, 1 H), 7.68 - 7.74 (m,
1 H), 7.97 - 8.04 (m, 1 H), 8.29 - 8.40 (m, 1 H), 8.60 - 8.65 (m, 1 H), 9.47 (br. s, 1 H); m/z (ES+APCI)+: 385 [M+H]+.
Example 65
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(pyridin-3-ylamino)-pyrimidin-4-ylamino]- propyl} -amide
Figure imgf000155_0001
Prepared analogously to Example 64 from Intermediate 8 and 3-aminopyridine to give desired product as a white solid (22 mg, 31%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.44 - 0.50 (m, 2 H), 0.80 - 0.86 (m, 2 H), 1.43 - 1.51 (m, 1 H), 1.65 - 1.77 (m, 3 H), 1.78 - 1.92 (m, 1 H), 1.93 - 2.03 (m, 2 H), 2.04 - 2.17 (m, 2 H), 2.91 - 3.02 (m, 1 H), 3.10 - 3.18 (m, 2 H), 3.39 - 3.45 (m, 2 H), 6.89 - 6.94 (m, 1 H), 7.24 (dd, 7=8.5, 4.8 Hz, 1 H), 7.63 (s, 1 H), 7.67 - 7.73 (m, 1 H), 8.04 (dd, /=4.6, 1.4 Hz, 1 H), 8.23 (ddd, J=8.4, 2.6, 1.4 Hz, 1 H), 8.86 (d, 7=2.3 Hz, 1 H), 9.10 (br. s, 1 H); m/z (ES+APCI)+: 367 [M+H]+. Example 66
Cyclopentanecarboxylic acid {3-[5-cyclopropyl-2-(pyridin-3-ylamino)-pyrimidin-4-ylamino]- propylj-amide
Figure imgf000156_0001
Intermediate 9 (63 mg, 0.195 mmol), 3-aminopyridine (22 mg, 0.234 mmol), tris(dibenzylideneacetone)dipalladium(0) (11 mg, 0.012 mmol), xantphos (9 mg, 0.016 mmol) and sodium tert-butoxide (56 mg, 0.585 mmol) were combined with dioxane (3 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 100 0C for 18 hours, evaporated and eluted through a silica plug, then purified by preparative LCMS (low pH buffer). The resulting TFA salt was subjected to anion exchange chromatography using an Isolute-NH2 cartridge to give the product as a white solid (15.5 mg, 21%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.43 - 0.50 (m, 2 H), 0.79 - 0.86 (m, 2 H), 1.42 - 1.51 (m, 3 H), 1.54 - 1.65 (m, 4 H), 1.65 - 1.75 (m, 4 H), 2.46 - 2.55 (m, 1 H), 3.09 - 3.18 (m, 2 H), 3.39 - 3.47 (m, 2 H), 6.87 - 6.96 (m, 1 H), 7.24 (dd, ./=8.2, 4.6 Hz, 1 H), 7.63 (s, 1 H), 7.77 - 7.88 (m, 1 H), 8.04 (dd, 7=4.6, 1.4 Hz, 1 H), 8.23 (ddd, J=SA, 1.4, 1.3 Hz, 1 H), 8.86 (d, 7=2.3 Hz, 1 H), 9.12 (br. s, 1 H); m/z (ES+APCI)+: 381 [M+H]+.
Example 67
Cyclopentanecarboxylic acid {3-[2-(3-acetylamino-phenylamino)-5-cyclopropyl-pyrimidin-4- ylaminoj -propyl) -amide
Figure imgf000157_0001
To a solution of Intermediate 5 (70 mg, 0.186 mmol) in DMF (2 ml) was added cyclopentanecarboxylic acid (30 μl, 0.279 mmol), then HATU (113 mg, 0.298 mmol) and N1N- diisopropylethylamine (194 μl, 1.17 mmol). The reaction was stirred at room temperature for 90 minutes. The mixture was evaporated then eluted through a 1 g Isolute-NH2 cartridge with 9:1 DCM: methanol. The crude product was subjected to preparative LCMS (low pH buffer) followed by flash chromatography using a Biotage SP4 (gradient elution from 0-8% methanol in DCM). The resulting TFA salt was subjected to anion exchange chromatography using an Isolute-NH2 cartridge to give a white solid (36 mg, 44%). 1H NMR (400 MHz, DMSO-^6, 85 0C) δ ppm 0.52 - 0.60 (m, 2 H), 0.90 - 0.97 (m, 2 H), 1.44 - 1.53 (m, 2 H), 1.54 - 1.68 (m, 5 H), 1.68 - 1.80 (m, 4 H), 2.06 (s, 3 H), 2.42 - 2.57 (m, 1 H), 3.10 - 3.18 (m, 2 H), 3.50 - 3.58 (m, 2 H), 7.15 - 7.23 (m, 1 H), 7.23 - 7.32 (m, 2 H), 7.46 - 7.56 (m, 2 H), 7.94 - 7.98 (m, 1 H), 8.11 - 8.19 (m, 1 H), 9.69 - 9.80 (m, 2 H); m/z (ES+APCI)+: 437 [M+H]+.
Example 68 Cyclobutanecarboxylic acid {3-[2-(3-acetylamino-phenylamino)-5-cyclopropyl-pyrimidin-4- ylamino] -propyl) -amide
Figure imgf000158_0001
Prepared analogously to Example 69 from Intermediate 5 and cyclobutanecarboxylic acid to give the desired product as a white foam (12 mg, 13%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.42 - 0.50 (m, 2 H), 0.78 - 0.87 (m, 2 H), 1.42 - 1.51 (m, 1 H), 1.61 - 1.78 (m, 3 H), 1.80 - 1.91 (m, 1 H), 1.92 - 2.04 (m, 5 H), 2.03 - 2.17 (m, 2 H), 2.89 - 3.00 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.40 - 3.49 (m, 2 H), 6.79 - 6.85 (m, 1 H), 7.04 - 7.13 (m, 2 H), 7.37 - 7.41 (m, 1 H), 7.59 (s, 1 H), 7.64 - 7.70 (m, 1 H), 7.93 (br. s, 1 H), 8.87 (br. s, 1 H), 9.78 (br. s, 1 H); m/z (ES+ APCI)+: 423 [M+H]+.
Example 69 Thiophene-2-carboxylic acid {3-[2-(3-amino-phenylamino)-5-cyclopropyl-pyrimidin-4-ylamino]- propyl} -amide
Figure imgf000158_0002
A suspension of Example 31 (32.8 mg, 0.064 mmol) in 4M hydrogen chloride in dioxane (1 ml, 4 mmol) was stirred vigorously for 4 h at room temperature The reaction mixture was evaporated to dryness and purified by preparative LCMS (high pH buffer) to give an off-white solid (17 mg, 65%). 1H NMR (400 MHz, DMSO-ύfe) δ ppm 0.42 - 0.50 (m, 2 H), 0.79 - 0.86 (m, 2 H), 1.41 - 1.51 (m, 1 H), 1.78 - 1.89 (m, 2 H), 3.25 - 3.37 (m, 2 H), 3.45 - 3.54 (m, 2 H), 6.11 - 6.16 (m, 1 H), 6.81 - 6.89 (m, 2 H), 6.97 (br. s, 1 H), 7.01 - 7.16 (m, 2 H), 7.55 (s, 1 H), 7.69 - 7.77 (m, 2 H), 8.52 - 8.59 (m, 1 H), 8.76 (br. s, 1 H); m/z (ES+APCI)+: 409 [M+H]+.
Example 70
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-hydroxymethyl-phenylamino)-pyrimidin-4- ylamino]-propyl}-amide. xHCl salt.
Figure imgf000159_0001
Example 70 was prepared analogously to Example 40 from Intermediate 8 and 3- (hydroxymethyl)aniline to give a white solid (26 mg, 37%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.53 - 0.60 (m, 2 H), 0.82 - 0.94 (m, 2 H), 1.50 - 1.59 (m, 1 H), 1.65 - 1.78 (m, 3 H), 1.78 - 1.91 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.02 - 2.15 (m, 2 H), 2.89 - 3.01 (m, 1 H), 3.06 - 3.16 (m, 2 H), 3.44 - 3.54 (m, 2 H), 4.51 (s, 2 H), 7.06 - 7.10 (m, 1 H), 7.31 - 7.36 (m, 1 H), 7.39 - 7.45 (m, 1 H), 7.58 - 7.65 (m, 2 H), 7.71 - 7.84 (m, 1 H), 8.61 (br. s, 1 H), 10.35 (br. s, 1 H), 11.98 (br. s, 1 H); m/z (ES+APCI)+: 396 [M+H]+.
Example 71 3-{4-[3-(Cyclobutanecarbonyl-amino)-propylamino]-5-cyclopropyl-pyrimidin-2-ylamino}-benzoic acid hydrochloride salt.
Figure imgf000160_0001
Example 71 was prepared analogously to Example 40 from Intermediate 8 and 3-aminobenzioc acid to give a white solid (139 mg, 22%). 1H NMR (400 MHz, DMSO-Cf6) δ ppm 0.43 - 0.50 (m, 2 H), 0.78 - 0.85 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.64 - 1.76 (m, 3 H), 1.78 - 1.90 (m, 1 H), 1.91 - 2.01 (m, 2 H), 2.03 - 2.15 (m, 2 H), 2.89 - 2.99 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.43 - 3.49 (m, 2 H), 6.83 - 6.90 (m, 1 H), 7.27 - 7.33 (m, 1 H), 7.39 - 7.43 (m, 1 H), 7.62 (s, 1 H), 7.64 - 7.70 (m, 1 H), 7.86 (dd, 7=8.2, 1.4 Hz, 1 H), 8.56 - 8.58 (m, 1 H), 9.12 (br. s, 1 H); m/z (ES+APCI)+: 410 [M+H]+.
Example 72
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3, 5-difluoro-phenylamino)-pyrimidin-4- ylamino]-propyl}-amide. Hydrochloride salt
Figure imgf000160_0002
Example 72 was prepared analogously to Example 40 from Intermediate 8 and 3,5-difluoroaniline to give a white solid (16 mg, 22%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.55 - 0.62 (m, 2 H), 0.89 - 0.96 (m, 2 H), 1.52 - 1.61 (m, 1 H), 1.66 - 1.80 (m, 3 H), 1.81 - 1.91 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.03 - 2.15 (m, 2 H), 2.90 - 3.01 (m, 1 H), 3.08 - 3.16 (m, 2 H), 3.46 - 3.53 (m, 2 H), 6.95 - 7.04 (m, 1 H), 7.33 - 7.42 (m, 2 H), 7.69 (s, 1 H), 7.76 - 7.83 (m, 1 H), 8.72 (br. s, 1 H), 10.83 (br. s, 1 H); m/z (ES+APCI)+: 402.2 [M+H]+. Example 73
3-{4-[3-(Cyclobutanecarbonyl-amino)-propylamino]-5-cyclopropyl-pyrimidin-2-ylamino}-N-(2- methoxy-ethyl)-benzamide
Figure imgf000161_0001
To a solution of 2-methoxyethylamine (9 μl, 0.1 mmol) in DMF (1 ml) was added Example 71 (30 mg, 0.067 mmol), HATU (41 mg, 0.11 mmol) and diisopropylethylamine (70 μl, 0.4 mmol) and the resulting mixture was stirred at room temperature for 72 hours. The mixture was evaporated then eluted through a Ig Isolute-NH2 cartridge with 9:1 DCM: methanol eluent, then subjected to preparative LCMS (low pH buffer). The resulting TFA salt was subjected anion exchange chromatography using an Isolute-NH2 cartridge to give the title compound as a white solid (20 mg, 64%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.43 - 0.50 (m, 2 H), 0.79 - 0.86 (m, 2 H), 1.42 - 1.52 (m, 1 H), 1.63 - 1.77 (m, 3 H), 1.78 - 1.91 (m, 1 H), 1.91 - 2.01 (m, 2 H), 2.04 - 2.16 (m, 2 H), 2.89 - 2.99 (m, 1 H), 3.08 - 3.16 (m, 2 H), 3.26 (s, 3 H), 3.35 - 3.49 (m, 6 H), 6.81 - 6.89 (m, 1 H), 7.23 - 7.30 (m, 2 H), 7.61 (s, 1 H), 7.66 - 7.72 (m, 1 H), 7.77 - 7.81 (m, 1 H), 8.31 - 8.39 (m, 2 H), 9.04 (br. s, 1 H); m/z (ES+APCI)+: 467 [M+H]+.
Examples 74-82
Examples 74-82 were prepared analogously to example M5 (the general structure is shown below followed by the tabulated examples).
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0002
Example 83
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-l,2,4-triazol-l-ylmethyl-phenylamino)- pyrimidin-4-ylamino] -propyl} -amide
Figure imgf000164_0001
Example 83 was prepared analogously to Example 43 from Intermediate 8 and 3-(4H-l,2,4-triazol- 4-ylmethyl)aniline to give the title compound as a white solid (45 mg, 62%). 1H ΝMR (400 MHz, DMSO-c4) δ ppm 0.43 - 0.49 (m, 2 H), 0.79 - 0.85 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.65 - 1.77 (m, 3 H), 1.78 - 1.91 (m, 1 H), 1.92 - 2.01 (m, 2 H), 2.04 - 2.15 (m, 2 H), 2.90 - 2.99 (m, 1 H), 3.09 - 3.16 (m, 2 H), 3.38 - 3.45 (m, 2 H), 5.34 (s, 2 H), 6.69 - 6.73 (m, 1 H), 6.80 - 6.86 (m, 1 H), 7.15 - 7.21 (m, 1 H), 7.60 (s, 1 H), 7.62 - 7.71 (m, 2 H), 7.74 - 7.78 (m, 1 H), 7.97 (s, 1 H), 8.62 (s, 1 H), 8.96 (br. s, 1 H); m/z (ES+APCI)+: 447 [M+H]+.
Examples 84-87 Examples 84-87 were prepared in a similar manner to Example 83 (the general structure is shown below followed by the tabulated examples).
Figure imgf000165_0001
Figure imgf000165_0002
Figure imgf000166_0002
Example 88
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-oxo-2,3-dihydro-lH-isoindol-5-ylamino)- pyrimidin-4-ylamino]-propyl}-amide hydrochloride salt.
Figure imgf000166_0001
Example 88 analogously to Example 40 from Intermediate 8 and 6-amino-2,3-dihydro-lH- isoindol-1-one to give the desired product as a pale yellow solid (23 mg, 31%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.54 - 0.61 (m, 2 H), 0.87 - 0.95 (m, 2 H), 1.50 - 1.60 (m, 1 H), 1.65 - 1.78 (m, 3 H), 1.79 - 1.90 (m, 1 H), 1.90 - 2.00 (m, 2 H), 2.02 - 2.15 (m, 2 H), 2.88 - 3.00 (m, 1 H), 3.09 - 3.18 (m, 2 H), 3.44 - 3.53 (m, 2 H), 4.37 (s, 2 H), 7.57 - 7.68 (m, 3 H), 7.77 - 7.82 (m, 1 H), 8.06 (d, 7=1.4 Hz, 1 H), 8.62 - 8.75 (m, 2 H), 10.64 (br. s, 1 H); m/z (ES+APCI)+: 421 [M+H]+.
Example 89
3-{4-[3-(Cyclobutanecarbonyl-amino)-propylaminoJ-5-cyclopropyl-pyrimidin-2-ylamino}-N-(lH- imidazol-2-ylmethyl)-benzamide. Hydrochloride salt.
Figure imgf000167_0001
To a solution of C-(l//-Imidazol-2-yl)-methyIamine (6 mg, 0.13 mmol) in DMF (1 ml) was added Example 71 (40 mg, 0.089 mmol), HATU (36 mg, 0.14 mmol) and diisopropylethylamine (62 μl, 0.54 mmol). The reaction was stirred at room temperature for 18 hours. The reaction mixture was evaporated, triturated with methanol, dried to give the title compound as a white solid (6 mg, 18%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.43 - 0.49 (m, 2 H), 0.79 - 0.85 (m, 2 H), 1.43 - 1.51 (m, 1 H), 1.63 - 1.77 (m, 3 H), 1.78 - 1.90 (m, 1 H), 1.91 - 2.01 (m, 2 H), 2.04 - 2.15 (m, 2 H), 2.88 - 2.99 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.41 - 3.47 (m, 2 H), 4.46 (d, /=5.5 Hz, 2 H), 6.78 - 6.87 (m, 2 H), 6.98 - 7.03 (m, 1 H), 7.25 - 7.30 (m, 1 H), 7.33 - 7.38 (m, 1 H), 7.61 (s, 1 H), 7.68 - 7.73 (m, 1 H), 7.80 - 7.84 (m, 1 H), 8.36 - 8.39 (m, 1 H), 8.76 - 8.81 (m, 1 H), 9.01 - 9.08 (m, 1 H), 11.76 (br. s, 1 H); m/z (ES+APCI)+: 489 [M+H]\
Example 90
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(l-oxo-2,3-dihydro-lH-isoindol-5-ylamino)- pyrimidin-4-ylamino]-propyl}-amide. Trifluoroacetic acid salt.
Figure imgf000168_0001
Intermediate 8 (50 mg, 0.162 mmol), 5-amino-2,3-dihydro-isoindol-l-one (72 mg, 0.485 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 150 0C for 60 minutes in a Biotage 1-60 microwave reactor. The reaction was evaporated, purified by LCMS (low pH buffer) triturated with methanol and dried to give the title compound as a white solid (6 mg, 7%). 1H NMR (400 MHz, DMSOd6) δ ppm 0.55 - 0.61 (m, 2 H), 0.87 - 0.93 (m, 2 H), 1.50 - 1.57 (m, 1 H), 1.64 - 1.77 (m, 3 H), 1.78 - 1.90 (m, 1 H), 1.91 - 2.00 (m, 2 H), 2.02 - 2.13 (m,
2 H), 2.88 - 2.98 (m, 1 H), 3.09 - 3.16 (m, 2 H), 3.28 - 3.56 (m, 2 H), 4.38 (s, 2 H), 7.61 - 7.69 (m,
3 H), 7.71 - 7.77 (m, 1 H), 7.86 - 7.91 (m, 1 H), 8.37 - 8.53 (m, 2 H), 10.39 (br. s, 1 H); m/z (ES+APCI)+: 421 [M+H]+.
Example 91
Cyclobutanecarboxylic acid (3-{5-cyclopropyI-2-[3-(4-methyI-piperazin-l-yl)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide. Trifluoroacetic acid salt.
Figure imgf000169_0001
Intermediate 8 (40 mg, 0.130 mmol), 3-(4-methyl-piperazin-l-yl)-phenylamine (30 mg, 0.155 mmol), Pd2(dba)3 (7 mg, 0.008 mmol), xantphos (6 mg, 0.01 mmol) and sodium tert-butoxide (37 mg, 0.385 mmol) were combined with dioxane (1 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 90 0C for 18 hours, evaporated and purified through a silica plug, eluting with 0 to 10% methanol/DCM. The crude product was further purified by LCMS to give the desired product as a white solid (24 mg, 32%). 1H NMR (400 MHz, DMS0-<4 850C) δ ppm 0.54 - 0.61 (m, 2 H), 0.90 - 0.97 (m, 2 H), 1.53 - 1.63 (m, 1 H), 1.70 - 1.94 (m, 4 H), 1.98 - 2.19 (m, 4 H), 2.88 (s, 3 H), 2.92 - 3.02 (m, 1 H), 3.11 - 3.18 (m, 2 H), 3.24 - 3.55 (m, 10 H), 6.80 (dd, J=8.0, 2.1 Hz, 1 H), 7.11 (dd, 7=8.0, 1.6 Hz, 1 H), 7.16 - 7.20 (m, 1 H), 7.24 - 7.31 (m, 1 H), 7.44 (br. s, 1 H), 7.55 (s, 1 H), 8.12 (br. s, 1 H), 9.82 (br. s, 1 H); m/z (ES+APCI)+: 464 [M+H]+.
Example 92
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-β-morpholin-4-ylmethyl-phenylamino)- pyrimidin-4-ylamino]-propyl}-amide. Trifluoroacetic acid salt.
Figure imgf000170_0001
Example 92 was prepared in a similar manner to Example 91 from Intermediate 8 and 3- (morpholin-4-ylmethyl)aniline to give the desired product as a white solid (21 mg, 28%). 1H NMR (400 MHz, DMSO-^6, 850C) δ ppm 0.56 - 0.62 (m, 2 H), 0.91 - 0.97 (m, 2 H), 1.55 - 1.63 (m, 1 H), 1.71 - 1.83 (m, 3 H), 1.83 - 1.94 (m, 1 H), 1.98 - 2.18 (m, 4 H), 2.92 - 3.02 (m, 1 H), 3.03 - 3.18 (m, 6 H), 3.47 - 3.54 (m, 2 H), 3.74 - 3.84 (m, 4 H), 4.18 - 4.24 (m, 2 H), 7.24 - 7.29 (m, 1 H), 7.43 - 7.50 (m, 2 H), 7.60 (s, 1 H), 7.63 - 7.65 (m, 1 H), 7.65 - 7.70 (m, 1 H), 8.22 (br. s, 1 H), 10.16 (br. s, 1 H); m/z (ES+ APCI)+: 465 [M+H]+.
Example 93
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-morpholin-4-yl-pyridin-3-ylcanino)-pyrimidin- 4-ylamino]-propyl}-amide
Figure imgf000171_0001
Intermediate 8 (40 mg, 0.130 mmol), 6-moφholin-4-yl-pyridin-3-ylamine (28 mg, 0.155 mmol), Pd2(dba)3 (7 mg, 0.008 mmol), xantphos (6 mg, 0.01 mmol) and sodium te/Y-butoxide (37 mg, 0.385 mmol) were combined with dioxane (1 ml), sealed and then purged with nitrogen gas. The reaction mixture was heated at 90 0C for 18 hours, evaporated and purified through a silica plug, eluting with 0 to 10% methanol/DCM. The crude product was triturated with diethyl ether, filtered and dried to give the desired product as a white solid (10 mg, 18%). 1H NMR (400 MHz, DMSO- d6) δ ppm 0.40 - 0.48 (m, 2 H), 0.76 - 0.85 (m, 2 H), 1.39 - 1.50 (m, 1 H), 1.62 - 1.78 (m, 3 H), 1.79 - 1.92 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.04 - 2.16 (m, 2 H), 2.90 - 3.01 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.28 - 3.33 (m, 4 H), 3.35 - 3.42 (m, 2 H), 3.67 - 3.73 (m, 4 H), 6.73 - 6.82 (m, 2 H), 7.56 (s, 1 H), 7.63 - 7.71 (m, 1 H), 7.97 (dd, /=9.2, 2.7 Hz, 1 H), 8.42 - 8.46 (m, 1 H), 8.67 (br. s, 1 H); m/z (ES+APCI)+: 452 [M+H]+.
Example 94 Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-f3-(2H-tetrazol-5-yl)-phenylaminoJ-pyrimidin-4- ylamino}-propyl)-aτnide
Figure imgf000172_0001
Intermediate 8 (50 mg, 0.162 mmol), 3-(2H-tetrazol-5-yl)-phenylamine (78 mg, 0.484 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The mixture was evaporated and purified by flash chromatography on the Biotage SP4, eluting with 0 to 12% methanol/DCM. The product was then eluted through a 0.5 g Isolute-NΗ2 cartridge with 9:1 DCM: methanol to give the free base as a white solid (16 mg, 23%). 1H NMR (400 MHz, DMSO-^6, 850C) δ ppm 0.46 - 0.50 (m, 2 H), 0.82 - 0.87 (m, 2 H), 1.47 - 1.55 (m, 1 H), 1.68 - 1.87 (m, 4 H), 1.92 - 2.01 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.89 - 2.98 (m, 1 H), 3.15 - 3.21 (m, 2 H), 3.53 - 3.60 (m, 2 H), 6.41 - 6.47 (m, 1 H), 7.15 - 7.21 (m, 1 H), 7.49 (dd, J=7.6, 1.6 Hz, 1 H), 7.50 - 7.53 (m, 1 H), 7.61 (s, 1 H), 7.65 - 7.71 (m, 1 H), 8.45 (br. s, 1 H), 8.55 - 8.58 (m, 1 H); m/z (ES+APCI)': 432 [M-H]\
Example 95
(3-{4-[3-(Cyclobutanecarbonyl-amino)-propylamino]-5-cyclopropyl-pyrimidin-2-ylamino}- phenyl) -acetic acid. Hydrochloride.
Figure imgf000173_0001
Intermediate 8 (50 mg, 0.162 mmol), (3-amino-phenyl)-acetic acid (74 mg, 0.484 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The mixture was evaporated and purified by flash chromatography on the Biotage SP4, eluting with 0 to 12% methanol/DCM to give the product as a white solid (54 mg, 73%). 1H NMR (400 MHz, DMSO-J6, 850C) δ ppm 0.53 - 0.59 (m, 2 H), 0.88 - 0.96 (m, 2 H), 1.53 - 1.63 (m, 1 H), 1.72 - 1.94 (m, 4 H), 1.99 - 2.08 (m, 2 H), 2.09 - 2.19 (m, 2 H), 2.90 - 3.21 (m, 3 H), 3.47 - 3.59 (m, 4 H), 6.94 - 7.02 (m, 1 H), 7.24 - 7.32 (m, 1 H), 7.42 - 7.52 (m, 2 H), 7.56 - 7.64 (m, 2 H), 7.86 (br. s, 1 H), 9.75 (br. s, 1 H); m/z (ES+APCI)+: 424 [M+H]+.
Example 96
Cyclobutanecarboxylic acid {3-[2-(3H-benzimidazol-5-ylamino)-5-cyclopropyl-pyrimidin-4- ylamino]-propyl}-amide
Figure imgf000174_0001
Intermediate 8 (50 mg, 0.162 mmol), 3H-benzimidazol-5-ylamine (65 mg, 0.484 mmol), glacial acetic acid (9 μl, 0.162 mmol) and n-butanol (1.5 ml) were combined and irradiated at 150 0C for 40 minutes in a Biotage 1-60 microwave reactor. The mixture was evaporated and purified by flash chromatography on the Biotage SP4, eluting with 0 to 12% methanol/DCM. The product was then eluted through a 0.5g Isolute-NΗ2 cartridge with 9:1 DCM: methanol. Further purification by LCMS (low pH buffer) gave the title compound as a white solid (13 mg, 15%). 1H NMR (400 MHz, DMSO-ck) δ ppm 0.58 - 0.63 (m, 2 H), 0.88 - 0.94 (m, 2 H), 1.52 - 1.60 (m, 1 H), 1.64 - 2.00 (m, 7 H), 2.02 - 2.13 (m, 2 H), 2.90 - 3.00 (m, 1 H), 3.10 - 3.17 (m, 2 H), 3.45 - 3.56 (m, 2 H), 7.61 (dd, J=SJ, 1.8 Hz, 1 H), 7.68 (s, 1 H), 7.80 - 7.87 (m, 2 H), 8.23 (d, J=IA Hz, 1 H), 8.56 - 8.62 (m, 1 H), 9.33 (s, 1 H), 10.94 (br. s, 1 H); m/z (ES+APCI)+: 406 [M+H]+.
Example 97
Cyclobutanecarboxylic acid {3-[2-(6-bromo-pyridin-3-ylamino)-5-cyclopropyl-pyrimidin-4- ylamino] '-propyl} -amide
Figure imgf000174_0002
A mixture of Intermediate 8 (200 mg, 0.649 mmol), 2-bromo-5-aminopyridine (225 mg, 1.30 mmol) and AcOH (37 μl, 0.649 mmol) in nBuOH were charged into a microwave reactor vial and sealed. The vial was placed in the Biotage 1-60 microwave reactor and irradiated at 1500C for 40 minutes. After the reaction a precipitate was observed. The mixture was filtered and the collected solid was washed with diethyl ether. The solid was dispersed into DCM and enough MeOH was added to give a clear solution. The solution was then washed with saturated aqueous sodium carbonate solution. The organic phase was dried and concentrated to give an off-white solid (214 mg, 74 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.44 - 0.50 (2 H, m), 0.79 - 0.86 (2 H, m), 1.43 - 1.51 (1 H, m), 1.63 - 1.77 (3 H, m), 1.81 - 2.02 (3 H, m), 2.04 - 2.16 (2 H, m), 2.92 - 3.02 (1 H, m), 3.05 - 3.17 (2 H, m), 3.37 - 3.45 (2 H, m), 6.96 (1 H, t, J=5.72 Hz), 7.48 (1 H, d, 7=9.16 Hz), 7.62 - 7.72 (2 H, m), 8.16 (1 H, dd, 7=8.70, 2.75 Hz), 8.76 (1 H, d, J=2.75 Hz), 9.29 (1 H, s); m/z (ES+APCI)+ : 445/447 [M+H]+.
Example 98
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-iodo-pyridin-3-ylamino)-pyrimidin-4- ylamino]-propyl}-amide
Figure imgf000175_0001
Prepared analogously to Example 97 from Intermediate 8 and 2-iodo-5-aminopyridine to give an off-white solid (209 mg, 65%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.42 - 0.50 (2 H, m), 0.77 - 0.86 (2 H, m), 1.43 - 1.51 (1 H, m), 1.62 - 1.77 (3 H, m), 1.82 - 2.02 (3 H, m), 2.04 - 2.16 (2 H, m), 2.97 (1 H, m), 3.12 (2 H, m), 3.37 - 3.45 (2 H, m), 6.95 (1 H, t, 7=5.95 Hz), 7.57 - 7.72 (3 H, m), 7.97 (1 H, dd, J=8.70, 2.75 Hz), 8.76 (1 H, d, 7=2.75 Hz), 9.25 (1 H, s); m/z (ES+APCI)+ : 493 [M+H]+.
Example 99
Cyclobutanecarboxylic acid {3-[2-(6-chloro-pyridin-3-ylamino)-5-cyclopropyl-pyrimidin-4- ylamino] -propyl} -amide
Figure imgf000176_0001
Prepared analogously to Example 97 from Intermediate 8 and 2-chloro-5-aminopyridine to give a mauve coloured solid (888 mg, 61%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.43 - 0.51 (2 H, m), 0.78 - 0.88 (2 H, m), 1.44 - 1.52 (1 H, m), 1.64 - 1.78 (3 H, m), 1.80 - 2.03 (3 H, m), 2.05 - 2.17 (2 H, m), 2.98 (1 H, m), 3.07 - 3.24 (2 H, m), 3.38 - 3.46 (2 H, m), 6.97 (1 H, t, 7=5.72 Hz), 7.37 (1 H, d, 7=8.70 Hz), 7.61 - 7.74 (2 H, m), 8.26 (1 H, dd, 7=8.93, 2.98 Hz), 8.77 (1 H, d, 7=2.75 Hz), 9.30 (1 H, s); m/z (ES+APCI)+ : 401/403 [M+H]+.
Example 100
Cyclobutanecarboxylic acid [3-(5-cyclopropyl-2-{6-[(2-dimethylamino-ethyl) -methyl-amino) '- pyridin-3-ylamino}-pyrimidin-4-ylamino)-propyl]-amide
Figure imgf000176_0002
Prepared analogously to Example 97 from intermediate 8 and Intermediate 13 to give a purple coloured foamy solid (18 mg, 24%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.39 - 0.49 (2 H, m), 0.76 - 0.84 (2 H, m), 1.40 - 1.47 (1 H, m), 1.60 - 1.77 (3 H, m), 1.82 - 1.90 (1 H, m), 1.91 - 2.02 (2 H, m), 2.04 - 2.20 (8 H, m), 2.36 (2 H, t, 7=7.10 Hz), 2.81 - 3.05 (4 H, m), 3.07 - 3.16 (2 H, m), 3.33 - 3.42 (2 H, m), 3.55 (2 H, t, 7=6.87 Hz), 6.52 (1 H, d, 7=9.16 Hz), 6.74 (1 H, t, 7=5.72 Hz), 7.53 (1 H, s), 7.69 (1 H, t, 7=5.72 Hz), 7.86 (1 H, dd, 7=9.16, 2.75 Hz), 8.30 (1 H, d, 7=2.29 Hz), 8.50 (1 H, s); m/z (ES+APCI)+ : 467 [M+H]+.
Example 101
Cyclobutanecarboxylic acid [3-(5-cyclopropyl-2-{4-[(2-hydroxy-ethyl)-methyl-amino]- phenylamino}-pyrimidin-4-ylamino)-propyl]-amide
Figure imgf000177_0001
Prepared analogously to Example 97 from Intermediate 8 and Intermediate 15 to give a purple coloured foamy solid (36 mg, 50 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.40 - 0.48 (2 H, m), 0.77 - 0.86 (2 H, m), 1.39 - 1.47 (1 H, m), 1.61 - 1.77 (3 H, m), 1.79 - 1.91 (1 H, m), 1.92 - 2.02 (2 H, m), 2.05 - 2.16 (2 H, m), 2.91 - 3.06 (4 H, m), 3.06 - 3.15 (2 H, m), 3.30 - 3.42 (2 H, m), 3.47 - 3.58 (4 H, m), 4.66 - 4.71 (1 H, m), 6.54 (1 H, d, ./=9.16 Hz), 6.73 (1 H, t, J=5.95 Hz), 7.53 (1 H, s), 7.67 (1 H, t, J=5.72 Hz), 7.85 (1 H, dd, ./=9.16, 2.75 Hz), 8.29 (1 H, d, J=2.75 Hz), 8.48 (1 H, s); m/z (ES+ APCI)+ : 441 [M+H]+.
Example 102
Cyclobutanecarboxylic acid [3-(5-cyclopropyl-2-{6-[methyl-(2-morpholin-4-yl-ethyl)-amino]- pyridin-3-ylamino}-pyrimidin-4-ylarnino)-propyl] -amide
Figure imgf000177_0002
Prepared analogously to Example 97 from Intermediate 8 and Intermediate 17 to give a purple coloured foamy solid (38 mg, 46 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.40 - 0.48 (2 H, m), 0.76 - 0.86 (2 H, m), 1.39 - 1.47 (1 H, m), 1.59 - 1.77 (3 H, m), 1.79 - 2.02 (3 H, m), 2.04 - 2.16 (2 H, m), 2.37 - 2.47 (6 H, m), 2.89 - 3.00 (4 H, m), 3.06 - 3.15 (2H, m), 3.34 - 3.43 (2 H, m), 3.48 - 3.65 (6 H, m), 6.53 (1 H, d, J=9.16 Hz), 6.72 (1 H, t, J=5.95 Hz), 7.53 (1 H, s), 7.66 (1 H, t, J=5.72 Hz), 7.86 (1 H, dd, 7=9.16, 2.75 Hz), 8.30 (1 H, d, J=2.29 Hz), 8.48 (1 H, s); m/z (ES+APCI)+ : 510 [M+H]+.
Example 103
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-hydroxymethyl-pyridin-3-ylamino)-pyrimidin- 4-ylamino]-propyl}-amide
Figure imgf000178_0001
A solution of Intermediate 21 (590 mg, 1.16 mmol) and tetrabutylammonium fluoride trihydrate (1.1 g, 3.47 mmol) in THF (40 ml) was stirred at rt for 90 minutes. The mixture was then concentrated to dryness and the residue was diluted with DCM. The DCM extract was washed with water, dried and concentrated. The residue was purified by trituration with diethyl ether twice, successively to afford a pale brown coloured solid (446 mg, 97 %). 1H NMR (400 MHz, DMSO- d6) δ ppm 0.44 - 0.50 (2 H, m), 0.79 - 0.87 (2 H, m), 1.43 - 1.51 (1 H, m), 1.65 - 1.78 (3 H, m), 1.82 - 1.92 (1 H, m), 1.93 - 2.03 (2 H, m), 2.06 - 2.16 (2 H, m), 2.93 - 3.02 (1 H, m), 3.09 - 3.19 (2 H, m), 3.42 (2 H, q, 7=6.41 Hz), 4.47 (2 H, d, /=5.50 Hz), 5.23 (1 H, t, 7=5.72 Hz), 6.88 (1 H, t, 7=5.95 Hz), 7.31 (1 H, d, 7=8.70 Hz), 7.62 (1 H, s), 7.69 (1 H, t, J=5.72 Hz), 8.18 (1 H, dd, 7=8.47, 2.52 Hz), 8.80 (1 H, d, 7=2.29 Hz), 9.04 (1 H, s); m/z (ES+APCI)+ : 397 [M+H]+.
Example 104
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-morpholin-4-ylmethyl-pyridin-3-ylamino)- pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000178_0002
A mixture of Intermediate 22 (30 mg), morpholine (8.2 μl, 0.095 mmol) and DIPEA (66 μl, 0.379 mmol) in DCM (2 ml) were stirred at rt over the weekend. The mixture was then diluted with DCM and washed with saturated aqueous sodium hydrogen carbonate solution. The organic phase was dried and concentrated. The crude residue was purified by flash column chromatography on silica gel (3 g) eluting with 10:1 DCM:MeOH to give a white solid (14 mg). 1H NMR (400 MHz,
DMSO-^6) δ ppm 0.44 - 0.49 (2 H, m), 0.80 - 0.86 (2 H, m), 1.43 - 1.50 (1 H, m), 1.65 - 1.77 (3 H, m), 1.81 - 1.89 (1 H, m), 1.93 - 2.02 (2 H, m), 2.05 - 2.16 (2 H, m), 2.32 - 2.43 (4 H, m), 2.93 - 3.02 (1 H, m), 3.09 - 3.17 (2 H, m), 3.37 - 3.50 (4 H, m), 3.52 - 3.60 (4 H, m), 6.88 (1 H, t, 7=5.95
Hz), 7.27 (1 H, d, 7=8.24 Hz), 7.62 (1 H, s), 7.69 (1 H, t, J=5.72 Hz), 8.18 (1 H, dd, 7=8.47, 2.52 Hz), 8.78 (1 H, d, 7=2.29 Hz), 9.05 (1 H, s); m/z (ES+APCI)+ : 466 [M+H]+.
Example 105
{3-[5-Cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-yloxy]-propyl}-carbamic acid tert-butyl ester
Figure imgf000179_0001
To solution of Intermediate 25 (24 mg, 0.071 mmol) in DMF (1 ml) was sequentially added added cyclobutanecarboxylic acid (10.2 μl, 0.11 mmol), HATU (43 mg, 0.11 mmol) and NN- diisopropylethylamine (74 μl, 0.43 mmol) and the reaction mixture left to stir at room temperature overnight. The solvents were removed under reduced pressure and the crude material re-dissolved 1 :9 methanol - DCM and eluted through an Isolute-ΝH2 cartridge. The solvents were removed and the crude product purified by preparative LCMS (high pH buffer) to give the product as a white solid (7 mg, 26%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.65 - 0.70 (m, 2 H), 0.75 - 0.85 (m, 2 H), 1.69 - 1.81 (m, 2 H), 1.83 - 2.01 (m, 5 H), 2.03 - 2.14 (m, 2 H), 2.91 - 3.01 (m, 1 H), 3.15 - 3.29 (m, 2 H), 4.37 (t, 7=6.18 Hz, 2 H), 6.70 (td, 7=8.36, 2.52 Hz, 1 H), 7.23 - 7.30 (m, 1 H), 7.43 - 7.48 (m, 1 H), 7.70 - 7.79 (m, 2 H), 7.94 (s, 1 H), 9.61 (s, 1 H); m/z (ES+APCI)+ : 407 [M+Na]+.
Example 106
Cyclobutanecarboxylic acid (3-{[5-cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-yl]-methyl- aminoj-propyl) -amide
Figure imgf000180_0001
Intermediate 27 (40 mg, 0.12 mmol), 3-fluoroaniline (14 μl, 0.15 mmol), Pd2(dba)3 (6.8 mg, 0.007 mmol), xantphos (5.8 mg, 0.01 mmol) and sodium ter/-butoxide (35.8 mg, 0.37 mmol) were combined in dioxane (2 ml), under nitrogen and the reaction mixture stirred at 90 0C for 2 days. The solvents were removed and the crude material re-dissolved in 1 :9 methanol - DCM and eluted through a silica plug. The solvents were removed and the crude product purified by preparative LCMS (low pH buffer). After solvent removal, the product was re-dissolved in methanol/DCM (1 :9) and eluted through an Isolute-NH2 cartridge and solvents evaporated to give an off-white solid (11 mg, 22%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.57 - 0.62 (m, 2 H), 0.78 - 0.87 (m, 2 H), 1.67 - 1.77 (m, 3 H), 1.79 - 1.90 (m, 2 H), 1.91 - 2.00 (m, 2 H), 2.02 - 2.14 (m, 2 H), 2.89 - 2.96 (m, 1 H), 3.01 - 3.13 (m, 2 H), 3.20 (s, 3 H), 3.67 (t, ./=7.33 Hz, 2 H), 6.64 (td, J=8.36, 2.52 Hz, 1 H), 7.19 - 7.25 (m, 1 H), 7.41 (d, /=8.24 Hz, 1 H), 7.68 (t, J=5.50 Hz, 1 H), 7.79 (dt, 7=12.82, 2.29 Hz, 1 H), 7.83 (s, 1 H), 9.24 (s, 1 H); m/z (ES+APCI)+: 398 [M+H]+.
Example 107
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylamino]- propylj-methyl-amide
Figure imgf000181_0001
To a stirred solution of Intermediate 29 (21 mg, 0.06 mmol) in DMF (1 ml) was added sequentially, cyclobutanecarboxylic acid (8.6 μl, 0.09 mmol), HATU (36 mg, 0.1 mmol) and N,N- diisopropylethylamine (62 μl, 0.4 mmol), and the reaction mixture was left to stir overnight at room temperature. The solvent was evaporated and the crude material was re-dissolved in 1 :9 methanol/DCM and eluted through an Isolute-NH2 cartridge. The crude material was purified by preparative LCMS (low pH buffer) and solvents removed. The product as its TFA salt was then eluted through an Isolute-NH2 cartridge using DCM:methanol (9:1), to give the final product as a white solid, (8 mg, 33%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.47 - 0.58 (m, 2 H), 0.82 - 0.95 (m, 2 H), 1.48 - 1.57 (m, 1 H), 1.67 - 1.83 (m, 3 H), 1.85 - 2.00 (m, 2 H), 2.07 - 2.23 (m, 4 H), 2.83 (s, 3 H), 3.20 - 3.36 (m, 1 H), 3.39 - 3.52 (m, 3 H), 6.63 - 6.70 (m, 1 H), 6.94 - 7.01 (m, 1 H), 7.22 - 7.29 (m, 1 H), 7.43 - 7.48 (m, 1 H), 7.69 (s, 1 H), 7.88 - 7.95 (m, 1 H), 9.21 (s, 1 H); m/z (ES+APCI)+: 398 [M+H]+.
Example 108
N-{3-[5-Cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylamino]-propyl}-butyramide
Figure imgf000182_0001
Prepared analogously to Example 107 from
Intermediate 3 (35 mg, 0.10 mmol) and butyric acid (14 μl, 0.16 mmol) to give a white solid (26 mg, 67%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.43 - 0.65 (m, 2 H), 0.85 - 0.92 (m, 5 H), 1.48 - 1.60 (m, 3 H), 1.72 - 1.80 (m, 2 H), 2.08 (t, J=7.3 Hz, 2 H), 3.19 (q, 7=6.6 Hz, 2 H), 3.40 - 3.52 (m, 2 H), 6.67 (td, ./=8.5, 2.3 Hz, 1 H), 6.96 (t, 7=5.7 Hz, 1 H), 7.22 - 7.29 (m, 1 H), 7.47 (d, J=8.2 Hz, 1 H), 7.69 (s, 1 H), 7.85 - 7.94 (m, 2 H), 9.20 (s, 1 H); m/z (ES+APCI)+: 372 [M+H]+
Example 109
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-morpholin-4-yl-pyridin-3-ylamino)-pyrimidin- 4-ylamino]-propyl}-methyl-amide
Figure imgf000182_0002
To a solution of Intermediate 40 (27 mg, 0.06 mmol) in DMF (1 ml) was added butyric acid (9 μl, 0.1 mmol), followed by HATU (39 mg, 0.1 mmol) then N,N-diisopropylethylamine (67 μl, 0.4 mmol) and the resulting solution was left to stir at room temperature overnight. The volatiles were removed under reduced pressure and the crude product was re-dissolved in 1 :9 methanol :DCM (1 ml) and eluted though an Isolute-NH2 cartridge. The solvents were removed and the crude product purified by preparative LCMS (low pH buffer) and eluted through an Isolute-NH2 cartridge to give a solid (10 mg, 33%). 1H NMR (400 MHz, DMSO-cfe, 40 0C) δ ppm 0.41 - 0.58 (m, 2 H), 0.83 - 0.92 (m, 2 H), 1.47 - 1.54 (m, 1 H), 1.69 - 1.90 (m, 4 H), 1.91 - 2.04 (m, 1 H), 2.09 - 2.23 (m, 3 H),
2.82 (s, 1 H), 2.89 (s, 2 H), 3.24 - 3.28 (m, 1 H), 3.32 - 3.49 (m, 8 H), 3.72 - 3.77 (m, 4 H), 6.73 -
6.83 (m, 2 H), 7.60 - 7.62 (m, 1 H), 7.95 - 8.00 (m, 1 H), 8.49 - 8.52 (m, 1 H), 8.60 (s, 1 H); m/z (ES+APCI)+: 466 [M+H]+
Example 110 l-{3-[5-Cyclopropyl-2-(3-fluoro-phenylamino)-pyrirnidin-4-ylamino]-propyl}-piperidin-2-one
Figure imgf000183_0001
Intermediate 31 (71 mg, 0.2 mmol), 3-fluoroaniline (66.5 μl, 0.7 mmol) and glacial acetic acid (42.2 μl, 0.7 mmol) were mixed together in n-butanol (1 ml) and heated in the microwave at
150 °C for 1 hour. The solvents were evaporated and the crude material was purified by preparative
LCMS (low pH buffer). The product as its TFA salt was then eluted through an Isolute-NH2 cartridge using DCM:methanol 9:1 to give the final product as a white solid (31 mg, 35%). 1H
NMR (400 MHz, DMSOd6) δ ppm 0.44 - 0.59 (m, 2 H), 0.83 - 0.91 (m, 2 H), 1.47 - 1.54 (m, 1 H), 1.68 - 1.85 (m, 6 H), 2.27 (t, 7=6.2 Hz, 2 H), 3.23 - 3.35 (m, 2 H), 3.39 - 3.49 (m, 4 H), 6.67 (td,
7=8.2, 2.3 Hz, 1 H), 7.05 (t, ./=6.0 Hz, 1 H), 7.22 - 7.29 (m, 1 H), 7.43 - 7.48 (m, 1 H), 7.68 (s, 1
H), 7.90 (dt, /=12.8, 2.3 Hz, 1 H), 9.23 (s, 1 H); m/z (ES+APCI)+: 384 [M+H]+
Example 111 l-fS-fS-Cyclopropyl^-fS-fluoro-phenylaminoJ-pyrimidm^-ylcaninoJ-propylJS-methyl-lH- pyridin-2-one
Figure imgf000184_0001
Prepared analogously to Example 110 from Intermediate 33 (49 mg, 0.15 mmol) and 3- fluoroaniline (44 μl, 0.46 mmol) to give a white solid (35 mg, 57%). 1H NMR (400 MHz, DMSO- dβ) δ ppm 0.44 - 0.64 (m, 2 H), 0.84 - 0.94 (m, 2 H), 1.49 - 1.57 (m, 1 H), 1.97 - 2.07 (m, 5 H), 3.48 (q, ./=6.4 Hz, 2 H), 4.04 (t, 7=6.9 Hz, 2 H), 6.18 (t, 7=6.6 Hz, 1 H), 6.67 (td, /=8.5, 2.3 Hz, 1 H), 7.02 (t, J=5.7 Hz, 1 H), 7.22 - 7.30 (m, 1 H), 7.34 (d, 7=5.5 Hz, 1 H), 7.45 - 7.49 (m, 1 H), 7.56 - 7.60 (m, 1 H), 7.69 (s, 1 H), 7.87 - 7.92 (m, 1 H), 9.21 (s, 1 H); m/z (ES+APCI)+: 394 [M+H]+
Example 112 l-{3-[5-Cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylamino]-propyl}-3-methyl- imidazolidin-2-one
Figure imgf000184_0002
Prepared analogously to Example 110 from Intermediate 35 (80 mg, 0.26 mmol) and 3- fluoroaniline (75 μl, 0.78 mmol) to give a white solid (26 mg, 26%). 1H NMR (400 MHz, DMSO- d6) δ ppm 0.42 - 0.58 (m, 2 H), 0.82 - 0.90 (m, 2 H), 1.46 - 1.54 (m, 1 H), 1.75 - 1.84 (m, 2 H), 2.67 (s, 3 H), 3.17 - 3.36 (m, 6 H), 3.42 - 3.51 (m, 2 H), 6.65 (td, 7=8.1, 2.1 Hz, 1 H), 6.93 (t, J=6.0
Hz, 1 H), 7.21 - 7.28 (m, 1 H), 7.46 (d, J=8.2 Hz, 1 H), 7.67 (s, 1 H), 7.90 (dt, J=13.3, 2.3 Hz, 1 H), 9.20 (s, 1 H); m/z (ES+APCI)+: 385 [M+H]+
Example 113 l-fS-P-Cyclopropyl^-fS-fluoro-phenylaminoJ-pyrimidin^-ylaminoJ-propylJ-S.S-dimethyl- pyrrolidin-2-one
Figure imgf000185_0001
Prepared analogously to Example 110 from Intermediate 37 (78 mg, 0.24 mmol) and 3- fluoroaniline (70 μl, 0.73 mmol) to give a white solid (34 mg, 27%). 1H NMR (400 MHz, DMSO- Ck) δ ppm 0.57 - 0.72 (m, 2 H), 0.91 - 1.01 (m, 2 H), 1.05 (s, 6 H), 1.55 - 1.62 (m, 1 H), 1.79 - 1.90 (m, 4 H), 3.30 (t, 7=6.9 Hz, 4 H), 3.39 - 3.69 (m, 2 H), 6.97 - 7.03 (m, 1 H), 7.37 (d, J=92 Hz, 1 H), 7.43 - 7.50 (m, 1 H), 7.63 - 7.68 (m, 1 H), 7.68 (s, 1 H), 8.44 (br. s, 1 H), 10.37 (br. s, 1 H); m/z (ES+APCI)+: 398 [M+H]+
Example 114 l-{3-[5-Cyclopropyl-2-(3-fluoroψhenylamino)-pyrirnidin-4-ylamino]-propyl}-3-trifluoromethyl- lH-pyridin-2-one
Figure imgf000186_0001
Intermediate 39 (22 mg, 0.06 mmol), 3-fluoroaniline (17 μl, 0.18 mmol) and glacial acetic acid (10.8 μl, 0.19 mmol) were mixed together in n-butanol (0.5 ml) and heated in the microwave at 150 0C for 1 hour. The solvents were evaporated and the crude material was purified by preparative LCMS (high pH buffer) to give a white solid (13 mg, 50%). 1H NMR (400 MHz, DMSOcZ6) δ ppm 0.42 - 0.60 (m, 2 H), 0.84 - 0.90 (m, 2 H), 1.48 - 1.55 (m, 1 H), 2.02 - 2.10 (m, 2 H), 3.48 - 3.54 (m, 2 H), 4.10 (t, /=7.1 Hz, 2 H), 6.40 (t, ./=6.9 Hz, 1 H), 6.66 (td, 7=8.2, 2.3 Hz, 1 H), 6.96 (t, /=5.7 Hz, 1 H), 7.22 - 7.28 (m, 1 H), 7.47 (d, /=9.6 Hz, 1 H), 7.69 (s, 1 H), 7.88 (dt, /=12.8, 2.3 Hz, 1 H), 7.96 (d, /=6.0 Hz, 1 H), 8.09 (dd, /=6.6, 2.1 Hz, 1 H), 9.20 (s, 1 H); m/z (ES+APCI)+: 448 [M+H]+
Example 115
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(l-methyl-lH-pyrazol-4-yl)-pyridin-3- ylamino]-pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000187_0001
Prepared analogously to Example 114 from Intermediate 8 (25 mg, 0.08 mmol) and Intermediate 41 (42 mg, 0.24 mmol) to give the title compound (2.1 mg, 6%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.43 - 0.64 (m, 2 H), 0.81 - 0.96 (m, 2 H), 1.46 - 1.60 (m, 1 H), 1.62 - 1.81 (m, 3 H), 1.81 - 1.96 (m, 1 H), 1.96 - 2.09 (m, 2 H), 2.09 - 2.27 (m, 2 H), 2.89 - 3.10 (m, 1 H), 3.10 - 3.29 (m, 2 H), 3.44 - 3.56 (m, 2 H), 3.82 - 3.99 (m, 3 H), 6.93 (t, /=5.3 Hz, 1 H), 7.55 (d, J=8.2 Hz, 1 H), 7.67 (s, 1 H), 7.73 (t, 7=5.5 Hz, 1 H), 7.91 (s, 1 H), 8.17 (s, 1 H), 8.25 (d, /=8.7 Hz, 1 H), 8.84 (s, 1 H), 9.11 (s, 1 H); m/z (ES+APCI)+: 447 [M+H]+
Example 116
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-pyrimidin-5-yl-pyridin-3-ylamino)-pyrimidin- 4-ylamino]-propyl}-amide
Figure imgf000187_0002
Example 97 (109 mg, 0.24 mmol), pyrimidine-5-boronic acid (33 mg, 0.27 mmol), Pd(dppf)Cl2 (10 mg, 0.01 mmol) and 2M sodium carbonate (267 μl) were mixed together in dioxane (2 ml) and the solution degassed for 5 min.. The reaction mixture was then stirred under nitrogen at 100 0C for 18 h. The reaction mixture was partitioned between water and ethyl acetate, extracted three times with ethyl acetate, the combined organic extracts washed with brine, dried (MgSO4) and solvents removed. The crude product was purified by flash chromatography using a Biotage SP4 (DCM/methanol gradient) to give a white solid (35 mg, 32%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.46 - 0.62 (m, 2 H), 0.86 - 0.91 (m, 2 H), 1.49 - 1.57 (m, 1 H), 1.71 - 1,81 (m, 3 H), 1.83 - 1.91 (m, 1 H), 1.96 - 2.05 (m, 2 H), 2.09 - 2.19 (m, 2 H), 2.98 - 3.03 (m, 1 H), 3.20 (q, 7=6.4 Hz, 2 H), 3.45 - 3.52 (m, 2 H), 7.01 (t, 7=6.0 Hz, 1 H), 7.70 - 7.76 (m, 2 H), 8.08 (d, .7=8.7 Hz, 1 H), 8.47 (dd, 7=8.7, 2.3 Hz, 1 H), 9.07 (d, 7=2.3 Hz, 1 H), 9.19 (s, 1 H), 9.40 - 9.44 (m, 3 H); m/z (ES+APCI)+: 445 [M+H]+
Example 117 Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-pyrimidin-5-yl-phenylamino)-pyrimidin-4- ylaminoj-propylj-amide
Figure imgf000188_0001
Example 141 below (100 mg, 0.23 mmol), pyrimidine-5-boronic acid (31 mg, 0.25 mmol), Pd(dppf)Cl2 (9.2 mg, 0.01 mmol) and 2M sodium carbonate (248 μl, 0.50 mmol) were mixed together in dioxane (2 ml) and the solution degassed for 5 min.. The reaction mixture was then stirred at 100 0C under nitrogen overnight. The solvents were evaporated and the crude product was purified by flash chromatography using a Biotage SP4 (DCM/methanol gradient) to give a beige solid (50 mg, 50%). 1H NMR (400 MHz, DMS(W6) δ ppm 0.42 - 0.62 (m, 2 H), 0.84 - 0.90 (m, 2 H), 1.48 - 1.56 (m, 1 H), 1.66 - 1.81 (m, 3 H), 1.85 - 1.95 (m, 1 H), 1.97 - 2.06 (m, 2 H), 2.09 - 2.19
(m, 2 H), 2.95 - 3.04 (m, 1 H), 3.09 - 3.18 (m, 2 H), 3.47 (q, J=6.4 Hz, 2 H), 6.92 (t, 7=6.0 Hz, 1 H), 7.29 (d, 7=8.2 Hz, 1 H), 7.43 (t, 7=8.0 Hz, 1 H), 7.69 (s, 1 H), 7.72 (t, 7=5.7 Hz, 1 H), 7.85 (d, j=9.6 Hz, 1 H), 8.31 (s, 1 H), 9.09 (s, 2 H), 9.12 (s, 1 H), 9.23 (s, 1 H); m/z (ES+APCI)+: 444 [M+H]+
Example 118
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(lH-pyrazol-4-yl)-pyridin-3-ylamino]- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000189_0001
Example 97 (80 mg, 0.18 mmol), 4,4)5,5-tetramethyl-2-(lH-pyrazol-4-yl)-l,3,2-dioxaborolane (38 mg, 0.20 mmol), Pd(dppf)Cl2 (7 mg, 0.01 mmol) and 2M sodium carbonate (198 μl) were mixed together in dioxane (1 ml) and the solution degassed. The reaction mixture was then stirred at 100 0C under nitrogen overnight. The reaction mixture was cooled under nitrogen, and volatiles evaporated. The crude material was re-dissolved in methanol, eluted through a plug of silica (DCM/methanol) and solvents evaporated. The crude material was purified by preparative LCMS (low pH buffer) to give the final product as a white solid (7 mg, 9%). 1H NMR (400 MHz, DMSO- d6) δ ppm 0.50 - 0.70 (m, 2 H), 0.93 - 0.98 (m, 2 H), 1.55 - 1.62 (m, 1 H), 1.70 - 1.81 (m, 3 H), 1.82 - 1.90 (m, 1 H), 1.94 - 2.03 (m, 2 H), 2.07 - 2.17 (m, 2 H), 2.94 - 3.03 (m, 1 H), 3.11 - 3.34 (m, 2 H), 3.34 - 3.63 (m, 2 H, obscured by water peak), 7.63 (s, 1 H), 7.76 (t, 7=5.7 Hz, 1 H), 7.81 (d, 7=8.7 Hz, 1 H), 8.04 - 8.10 (m, 1 H), 8.23 (br. s, 2 H), 8.45 (br. s, 1 H), 8.73 (br. s, 1 H), 10.21 (br. s, 1 H); m/z (ES+APCI)+: 433 [M+H]+ Example 119
Cyclobutanecarboxylic acid {3-[2-([2,4']bipyridinyl-5-ylamino)-5-cyclopropyl-pyrimidin-4- ylamino] -propyl} -amide
Figure imgf000190_0001
Example 97 (80 mg, 0.18 mmol), 4-pyridineboronic acid pinacol ester (41 mg, 0.20 mmol), Pd(dppf)Cl2 (7 mg, 0.01 mmol) and 2M sodium carbonate (198 μl) were mixed together in dioxane (1 ml) and the solution degassed. The reaction mixture was then stirred under nitrogen at 100 0C overnight. The reaction mixture was cooled under nitrogen, and volatiles evaporated. The crude material was re-dissolved in methanol, eluted through a plug of silica (DCM/methanol) and solvents evaporated. The crude material was purified by preparative LCMS (low pH buffer). The product, as its TFA salt was eluted through an Isolute-NH2 cartridge using DCM:methanol 9:1 to give a yellow solid (16 mg, 20%). 1H NMR (400 MHz, DMSO-cfe) δ ppm 0.46 - 0.61 (m, 2 H), 0.82 - 0.94 (m, 2 H), 1.50 - 1.58 (m, 1 H), 1.69 - 1.94 (m, 4 H), 1.96 - 2.06 (m, 2 H), 2.10 - 2.21 (m, 2 H), 2.97 - 3.07 (m, 1 H), 3.15 - 3.25 (m, 2 H), 3.40 - 3.55 (m, 2 H), 7.03 (t, 7=6.0 Hz, 1 H), 7.72 (s, 1 H), 7.76 (t, 1 H), 8.02 - 8.05 (m, 2 H), 8.09 (d, 7=9.2 Hz, 1 H), 8.48 (dd, 7=8.9, 2.5 Hz, 1 H), 8.64 - 8.69 (m, 2 H), 9.06 (d, 7=2.3 Hz, 1 H), 9.47 (s, 1 H); m/z (ES+APCI)+: 444 [M+H]+
Example 120
Cyclobutanecarboxylic acid {3-[2-([2,3']bipyridinyl-5-ylamino)-5-cyclopropyl-pyrimidin-4- ylamino] -propyl} -amide
Figure imgf000191_0001
Example 97 (80 mg, 0.18 mmol), 3-pyridineboronic acid (24 mg, 0.20 mmol), Pd(dppf)Cl2 (7 mg, 0.01 mmol) and 2M sodium carbonate (198 μl) were mixed together in dioxane (1 ml) and the solution degassed. The reaction mixture was then stirred under nitrogen at 100 0C overnight. The reaction mixture was cooled under nitrogen, and volatiles evaporated. The crude material was re- dissolved in methanol, eluted through a plug of silica (DCM/methanol) and solvents evaporated. The crude material was purified by preparative LCMS (low pH buffer). The product, as its TFA salt was eluted through an Isolute-NH2 cartridge using DCM:methanol 9: 1 to give a white solid (34 mg, 43%).'H NMR (400 MHz, DMSO-^6) δ ppm 0.47 - 0.62 (m, 2 H), 0.84 - 0.91 (m, 2 H), 1.49 - 1.57 (m, 1 H), 1.70 - 1.91 (m, 4 H), 1.95 - 2.05 (m, 2 H), 2.09 - 2.20 (m, 2 H), 3.01 (quin, 7=8.5 Hz, 1 H), 3.20 (q, /=6.4 Hz, 2 H), 3.45 - 3.53 (m, 2 H), 7.00 (t, /=6.0 Hz, I H), 7.51 (dd, /=8.0, 4.8 Hz, 1 H), 7.70 (s, 1 H), 7.75 (t, /=5.7 Hz, 1 H), 8.00 (d, /=9.2 Hz, 1 H), 8.38 - 8.45 (m, 2 H), 8.57 - 8.59 (m, 1 H), 9.04 (d, /=2.7 Hz, 1 H), 9.25 (d, /=1.8 Hz, 1 H), 9.36 (s, 1 H); m/z (ES+APCI)+: 444 [M+H]+
Example 121
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-pyridin-4-yl-phenylamino)-pyrimidin-4- ylamino]-propyl}-amide
Figure imgf000192_0001
Example 141 (70 mg, 0.16 mmol), pyridine-4-boronic acid (22 mg, 0.17 mmol), Pd(dppf)Cl2 (6.4 mg, 0.01 mmol) and 2M sodium carbonate (173 μl, 0.35 mmol) were mixed together in dioxane (1.5 ml) and the solution degassed. The reaction mixture was then stirred at 100 0C under nitrogen overnight. The volatiles were evaporated and the crude product eluted through a plug of silica, followed by purification by preparative LCMS (low pH buffer). The product as its TFA salt was then eluted through an Isolute-NH2 cartridge using DCM:methanol 9:1, to give a white solid (37 mg, 53%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.40 - 0.61 (m, 2 H), 0.81 - 0.91 (m, 2 H), 1.47 - 1.55 (m, 1 H), 1.66 - 1.80 (m, 3 H), 1.82 - 1.94 (m, 1 H), 1.95 - 2.05 (m, 2 H), 2.07 - 2.18 (m, 2 H), 2.93 - 3.05 (m, 1 H), 3.13 (q, /=6.4 Hz, 2 H), 3.39 - 3.52 (m, 2 H), 6.90 (t, ./=6.0 Hz, 1 H), 7.27 (d, /=7.8 Hz, 1 H), 7.39 (t, /=7.8 Hz, 1 H), 7.63 - 7.66 (m, 2 H), 7.68 (s, 1 H), 7.70 (t, /=5.7 Hz, 1 H), 7.84 (d, /=8.2 Hz, 1 H), 8.31 (s, 1 H), 8.64 - 8.68 (m, 2 H), 9.10 (s, 1 H); m/z (ES+ APCI)+: 443 [M+H]+.
Example 122
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[3-(4-methyl-piperazin-l-ylmethyl)- phenylamino]-pyrimidin-4-ylamino} -propyl) -amide.
Figure imgf000193_0001
Intermediate 42 (125 mg, 0.27 mmol) and N-methyl piperazine (300 μl, 27 mmol) were combined in THF (2 ml) and stirred at room temperature for 18 h. The reaction mixture was diluted with EtOAc (20 ml) and partitioned with water (20 ml). The organic phase was washed with brine, dried (MgSO4) and evaporated. The crude material was purified by flash chromatography on the Biotage SP4, eluting with 0 to 10% Methanol/DCM gradient. Further purication by elution through a Ig Isolute-NH2 cartridge with 9:1 DCM: methanol eluent gave the desired product as a white solid (23%). 1H NMR (400 MHz, DMSO-*) δ ppm 0.43 - 0.49 (m, 2 H), 0.79 - 0.86 (m, 2 H), 1.42 - 1.52 (m, 1 H), 1.65 - 1.78 (m, 3 H), 1.78 - 1.92 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.04 - 2.17 (m, 5 H), 2.17 - 2.47 (m, 6 H), 2.89 - 3.00 (m, 1 H), 3.09 - 3.18 (m, 2 H), 3.32 - 3.38 (m, 4 H), 3.42 - 3.49 (m, 2 H), 6.73 - 6.78 (m, 1 H), 6.81 - 6.87 (m, 1 H), 7.08 - 7.17 (m, 1 H), 7.51 - 7.57 (m, 1 H), 7.60 (d, /=0.9 Hz, 1 H), 7.66 - 7.72 (m, 1 H), 7.80 - 7.84 (m, 1 H), 8.86 (br. s, 1 H); m/z (ES+APCI)+: 478 [M+H]+.
Example 123
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[3-(l-morpholin-4-yl-ethyl)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide.
Figure imgf000193_0002
Intermediate 8 (121 mg, 0.39 mmol), Intermediate 43 (122 mg, 0.59 mmol) and trifluoroacetic acid (29 μl, 0.39 mmol) were combined with n-butanol (1.5 ml) and heated at 75 0C for 1.5h The reaction mixture was evaporated and then purified by LCMS (low pH buffer). The product was then eluted through a 1 g Isolute-NH2 cartridge with 9:1 DCM: methanol to give a white solid (61%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.44 - 0.51 (m, 2 H), 0.81 - 0.87 (m, 2 H), 1.26 -
1.39 (m, 3 H), 1.44 - 1.52 (m, 1 H), 1.66 - 1.77 (m, 3 H), 1.78 - 1.92 (m, 1 H), 1.93 - 2.03 (m, 2 H), 2.04 - 2.16 (m, 2 H), 2.30 - 2.45 (m, 2 H), 2.90 - 3.01 (m, 1 H), 3.09 - 3.17 (m, 2 H), 3.21 - 3.63 (m, 9 H), 6.81 - 6.89 (m, 1 H), 7.05 (br. s, 1 H), 7.16 - 7.23 (m, 1 H), 7.54 - 7.58 (m, 1 H), 7.60 (d, .7=0.9 Hz, 1 H), 7.65 - 7.72 (m, 1 H), 7.77 - 7.81 (m, 1 H), 9.03 (br. s, 1 H); m/z (ES+APCI)+: 479 [M+H]+.
Example 124
Cyclobutanecarboxylic acid β-ft-cyclopropyl^-fS-φyrrolidine-l-carbonylj-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide.
Figure imgf000194_0001
Prepared in a similar manner to Example 73 from Example 71 and pyrrolidine, to give the desired product as a white solid (24 mg, 88%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.44 - 0.50 (m, 2 H), 0.78 - 0.87 (m, 2 H), 1.42 - 1.51 (m, 1 H), 1.62 - 1.91 (m, 8 H), 1.92 - 2.02 (m, 2 H), 2.04 - 2.16 (m, 2 H), 2.90 - 3.01 (m, 1 H), 3.07 - 3.16 (m, 2 H), 3.31 - 3.48 (m, 6 H), 6.85 - 6.90 (m, 1 H), 6.91 - 6.96 (m, 1 H), 7.22 - 7.28 (m, 1 H), 7.62 (s, 1 H), 7.68 - 7.73 (m, 1 H), 7.73 - 7.78 (m, 1 H), 8.00 - 8.04 (m, 1 H), 9.05 (br. s, 1 H); m/z (ES+APCI)+: 463 [M+H]+.
Examples 125-126
Examples 125-126 were prepared analogously to Example 124 (the general structure is shown below followed by the tabulated examples).
Figure imgf000194_0002
Figure imgf000194_0003
Figure imgf000195_0002
*HPLC column: 21.2xl00mm (5μm) C-18 Phenomenex Gemini; flow rate: 20ml/min; run time: 10 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: 0.1 % trifluoroacetic acid is added to the water.
Example 127
Cyclobutanecarboxylic acid [3-(5-cyclopropyl-2-{3-[2-(4-methyl-piperazin-l-yl)-2-oxo-ethyl]- phenylamino}-pyrimidin-4-ylamino)-propyl]-amide.
Figure imgf000195_0001
To a solution of N-methylpiperazine (8 μl, 0.07 mmol) in DMF (1 ml) was added Example 95 (50 mg, 0.11 mmol), HATU (44 mg, 0.12 mmol) and diisopropylethylamine (76 μl, 0.43 mmol), and the reaction was stirred at room temperature for 18 hours. The mixture was evaporated then eluted through a 0.5g Isolute-Nrh cartridge with 9:1 DCM: methanol eluent, then purified by LCMS (low pH buffer). The product was then eluted through a 0.5g Isolute-NH2 cartridge with 9:1 DCM: methanol to liberate the free base as a white solid (23 mg, 42%). 1H NMR (400 MHz, DMSO-Gf6) δ ppm 0.43 - 0.49 (m, 2 H), 0.78 - 0.86 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.64 - 1.77 (m, 3 H), 1.79 -
1.91 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.04 - 2.16 (m, 5 H), 2.17 - 2.26 (m, 4 H), 2.89 - 3.01 (m, 1 H), 3.08 - 3.16 (m, 2 H), 3.39 - 3.49 (m, 6 H), 3.63 (s, 2 H), 6.65 - 6.71 (m, 1 H), 6.78 - 6.85 (m, 1 H),
7.08 - 7.16 (m, 1 H), 7.53 - 7.58 (m, 1 H), 7.60 (s, 1 H), 7.65 - 7.73 (m, 2 H), 8.88 (br. s, 1 H); m/z (ES+APCI)+: 506 [M+H]+.
Example 128 Cyclobutanecarboxylic acid [3-(5-cyclopropyl-2-{3-[2-(4-dimethylamino-piperidin-l-yl)-2-oxo- ethyl]-phenylamino}-pyrimidin-4-ylamino)-propylj '-amide.
Figure imgf000196_0001
Example 128 was prepared in a similar manner to Example 127 from Example 95 and 4- (dimethylamino)piperidine to give the desired product as an off-white solid (20 mg, 56%). 1H
NMR (400 MHz, DMSO-^6) δ ppm 0.45 - 0.50 (m, 2 H), 0.82 - 0.88 (m, 2 H), 1.14 - 1.25 (m, 2 H),
1.46 - 1.54 (m, 1 H), 1.63 - 1.72 (m, 2 H), 1.72 - 1.93 (m, 4 H), 1.98 - 2.08 (m, 2 H), 2.08 - 2.19
(m, 8 H), 2.23 - 2.31 (m, 1 H), 2.92 - 3.02 (m, 1 H), 3.02 - 3.08 (m, 4 H), 3.14 - 3.22 (m, 2 H), 3.44
- 3.51 (m, 2 H), 3.64 (s, 2 H), 6.45 - 6.55 (m, 1 H), 6.68 - 6.76 (m, 1 H), 7.09 - 7.17 (m, 1 H), 7.38 (br. s, 1 H), 7.53 - 7.57 (m, 1 H), 7.61 (d, J=0.9 Hz, 1 H), 7.68 - 7.71 (m, 1 H), 8.47 (br. s, 1 H); m/z (ES+APCI)+: 534 [M+H]+.
Example 129
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(lH-indazol-6-ylamino)-pyrimidin-4-ylamino]- propylj-amide. Hydrochloride.
Figure imgf000197_0001
Prepared analogously to Example 40 from Intermediate 8 and 6-aminoindazole to give the desired product as a white solid (25 mg, 35%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.55 - 0.61 (m, 2 H), 0.88 - 0.94 (m, 2 H), 1.53 - 1.62 (m, 1 H), 1.64 - 1.74 (m, 1 H), 1.75 - 1.88 (m, 3 H), 1.89 - 1.99 (m, 2 H), 2.03 - 2.14 (m, 2 H), 2.91 - 3.01 (m, 1 H), 3.13 - 3.20 (m, 2 H), 3.50 - 3.57 (m, 2 H), 7.10 - 7.15 (m, 1 H), 7.66 (s, 1 H), 7.75 (d, 7=8.7 Hz, 1 H), 7.88 - 7.94 (m, 1 H), 8.01 - 8.06 (m, 2 H), 8.61 - 8.68 (m, 1 H), 10.64 (s, 1 H), 12.01 (br. s, 1 H); m/z (ES+APCI)+: 406 [M+H]+.
Example 130
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(lH-indazol-5-ylamino)-pyrimidin-4-ylamino]- propyl} -amide.
Figure imgf000197_0002
Prepared in a similar manner to Example 43 from Intermediate 8 and 5-aminoindazole. The product was isolated as a white solid (41 mg, 62%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.43 - 0.49 (m, 2 H), 0.78 - 0.86 (m, 2 H), 1.41 - 1.51 (m, 1 H), 1.67 - 1.78 (m, 3 H), 1.78 - 1.91 (m, 1 H), 1.91 - 2.02 (m, 2 H), 2.05 - 2.17 (m, 2 H), 2.89 - 3.01 (m, 1 H), 3.12 - 3.20 (m, 2 H), 3.40 - 3.49 (m, 2 H), 6.78 - 6.85 (m, 1 H), 7.38 (d, 7=9.2 Hz, 1 H), 7.54 (dd, 7=9.2, 1.8 Hz, 1 H), 7.61 (s, 1 H), 7.65 - 7.73 (m, 1 H), 7.93 (s, 1 H), 8.30 (d, 7=1.4 Hz, 1 H), 8.85 (s, 1 H); m/z (ES+APCI)+: 406 [M+H]+.
Example 131 Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-pyrrolidin-l-ylmethyl-phenylamino)- pyrimidin-4-ylamino]-propyl}-amide.
Figure imgf000198_0001
Intermediate 44 (40 mg, 0.089 mmol), pyrrolidine (0.13 mmol) and diisopropylethylamine (0.53 mmol) were combined in DCM (2 ml) and stirred at room temperature for 18 h and then heated at 350C for 30 min. The mixture was evaporated and the residue was eluted through a 0.5g Isolute- NH2 cartridge with 9:1 DCM: methanol, and then purified by flash chromatography on the Biotage SP4, eluting with 0 to 15% methanol/DCM gradient, to give the desired product as a white foam (12 mg, 29%). 1H NMR (400 MHz, DMS(W6) δ ppm 0.39 - 0.46 (m, 2 H), 0.75 - 0.83 (m, 2 H), 1.38 - 1.47 (m, 1 H), 1.59 - 1.73 (m, 7 H), 1.74 - 1.88 (m, 1 H), 1.89 - 1.99 (m, 2 H), 2.01 - 2.13 (m, 2 H), 2.32 - 2.41 (m, 4 H), 2.85 - 2.96 (m, 1 H), 3.06 - 3.15 (m, 2 H), 3.37 - 3.48 (m, 4 H), 6.71 - 6.75 (m, 1 H), 6.76 - 6.83 (m, 1 H), 7.05 - 7.13 (m, 1 H), 7.48 - 7.54 (m, 1 H), 7.57 (d, J=0.9 Hz, 1 H), 7.60 - 7.67 (m, 1 H), 7.79 (s, 1 H), 8.78 - 8.83 (m, 1 H); m/z (ES+APCI)+: 449 [M+H]+.
Example 132
Cyclobutanecarboxylic acid [3-(2-{3-[(cyclopentyl-methyl-amino)-methyl]-phenylamino}-5- cyclopropyl-pyrimidin-4-ylamino) -propyl] -amide.
Figure imgf000198_0002
Intermediate 44 (40 mg, 0.089 mmol), cyclopentylmethylamine (0.13 mmol) and diisopropylethylamine (0.53 mmol) were combined in DCM (2 ml) and stirred at room temperature for 18 h and then heated at 35 0C for 30 min. The mixture was evaporated and purified LCMS (low pH buffer). The product was then eluted through a 0.5 g Isolute-NH2 cartridge with 9:1 DCM: methanol to give the a white foam (12%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.43 - 0.49 (m, 2 H), 0.79 - 0.85 (m, 2 H), 1.35 - 1.56 (m, 5 H), 1.57 - 1.91 (m, 8 H), 1.92 - 2.04 (m, 5 H), 2.04 - 2.16 (m, 2 H), 2.65 - 2.74 (m, 1 H), 2.88 - 2.99 (m, 1 H), 3.09 - 3.16 (m, 2 H), 3.39 (s, 2 H), 3.42 - 3.49
(m, 2 H), 6.74 - 6.79 (m, I H), 6.79 - 6.84 (m, I H), 7.10 - 7.15 (m, 1 H), 7.53 - 7.57 (m, 1 H), 7.60 (d, J=0.9 Hz, 1 H), 7.64 - 7.69 (m, 1 H), 7.78 - 7.81 (m, 1 H), 8.84 (br. s, 1 H); m/z (ES+APCI)+: 477 [M+H]+.
Example 133
Cyclobutanecarboxylic acid [3-(5-cyclopropyl-2-{3-[(isobutyl-methyl-amino)-methyl]- phenylamino}-pyrimidin-4-ylamino)-propyl]-amide.
Figure imgf000199_0001
Prepared analogously to Example 131 from Intermediate 44 and isobutylmethylamine to give the desired product as a white foam (5.2 mg, 12%). 1H NMR (400 MHz, DMS0-<4 85 0C) δ ppm 0.43 - 0.51 (m, 2 H), 0.80 - 0.94 (m, 8 H), 1.46 - 1.55 (m, 1 H), 1.70 - 1.94 (m, 5 H), 1.98 - 2.08 (m, 2 H), 2.08 - 2.21 (m, 7 H), 2.91 - 3.13 (m, 1 H), 3.14 - 3.21 (m, 2 H), 3.40 (s, 2 H), 3.44 - 3.52 (m, 2 H), 6.46 - 6.53 (m, 1 H), 6.78 - 6.85 (m, 1 H), 7.10 - 7.18 (m, 1 H), 7.36 (br. s, 1 H), 7.56 - 7.63 (m, 2 H), 7.68 - 7.74 (m, 1 H), 8.44 (br. s, 1 H); m/z (ES+APCI)+: 465 [M+H]+.
Example 134
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3-piperidin-l-ylmethyl-phenylamino)-pyrimidin- 4-ylamino]-propyl}-amide.
Figure imgf000199_0002
Example 134 was prepared analogously to Example 131 from Intermediate 44 and piperidine to give the desired product as a white solid (21 mg, 51%). 1H NMR (400 MHz, DMSO-d6, 85 0C) δ ppm 0.44 - 0.50 (m, 2 H), 0.82 - 0.89 (m, 2 H), 1.35 - 1.57 (m, 7 H), 1.68 - 1.93 (m, 4 H), 1.97 -
2.20 (m, 4 H), 2.37 - 2.45 (m, 4 H), 2.90 - 3.12 (m, 1 H), 3.14 - 3.21 (m, 2 H), 3.37 - 3.55 (m, 4 H), 6.48 - 6.55 (m, 1 H), 6.79 - 6.84 (m, 1 H), 7.11 - 7.18 (m, 1 H), 7.36 (br. s, 1 H), 7.57 - 7.62 (m, 2 H), 7.72 - 7.78 (m, 1 H), 8.47 (br. s, 1 H); m/z (ES+APCI)+: 463 [M+H]+.
Example 135
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[3-(4, 4-difluoro-piperidin-l-ylmethyl)- phenylamino]-pyrimidin-4-ylamino} -propyl) -amide.
Figure imgf000200_0001
Example 135 was prepared analogously to Example 131 from Intermediate 44 and 4,4- difluoropiperidine to give the desired product as an off-white solid (33 mg, 75%).1H NMR (400 MHz, DMSO-<4) δ ppm 0.43 - 0.49 (m, 2 H), 0.79 - 0.86 (m, 2 H), 1.42 - 1.51 (m, 1 H), 1.65 - 1.77 (m, 3 H), 1.78 - 2.03 (m, 7 H), 2.05 - 2.16 (m, 2 H), 2.44 - 2.48 (m, 4 H), 2.88 - 3.01 (m, 1 H), 3.10 - 3.17 (m, 2 H), 3.42 - 3.49 (m, 4 H), 6.76 - 6.80 (m, 1 H), 6.81 - 6.86 (m, 1 H), 7.10 - 7.18 (m, 1 H), 7.57 - 7.59 (m, 1 H), 7.60 (d, J=0.9 Hz, 1 H), 7.65 - 7.70 (m, 1 H), 7.78 - 7.83 (m, 1 H), 8.87 (br. s, 1 H); m/z (ES+APCI)+: 499 [M+H]+.
Example 136
Cyclobutanecarboxylic acid {3-[2-(3-cyanomethyl-phenylamino)-5-cyclopropyl-pyrimidin-4- ylaminoj -propyl} -amide.
Figure imgf000200_0002
Intermediate 8 (324 mg, 1.05 mmol), Intermediate 45 (208 mg, 1.57 mmol) and trifluoroacetic acid (78 μl, 1.05 mmol) were heated in n-butanol (3 ml) at 750C for 3h. The resultant white precipitate was filtered, washing with a minimum amount of methanol. The solid was dissolved in 9: 1 DCM;methanol and eluted through a Ig Isolute-NH2 cartridge with 9:1 DCM: methanol to give the product as a white solid (0.33 g, 78%). 1H NMR (400 MHz, DMS(W6) δ ppm 0.46 - 0.52 (m, 2 H),
0.81 - 0.88 (m, 2 H), 1.45 - 1.54 (m, 1 H), 1.66 - 1.78 (m, 3 H), 1.79 - 1.92 (m, 1 H), 1.93 - 2.02 (m, 2 H), 2.04 - 2.17 (m, 2 H), 2.90 - 3.02 (m, 1 H), 3.08 - 3.18 (m, 2 H), 3.42 - 3.50 (m, 2 H), 3.99 (s, 2 H), 6.84 - 6.88 (m, 1 H), 7.18 - 7.22 (m, 1 H), 7.23 - 7.28 (m, 1 H), 7.57 - 7.61 (m, 1 H), 7.62 (d, /=0.9 Hz, 1 H), 7.67 - 7.74 (m, 1 H), 7.83 - 7.88 (m, 1 H), 9.28 (br. s, 1 H); m/z (ES+APCI)+: 405[M+H]+.
Example 137
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(l-methyl-lH-indazol-5-ylamino)-pyrimidin-4- ylamino]-propyl}-amide.
Figure imgf000201_0001
Example 137 was prepared analogously to Example 94 to from Intermediate 8 and 1 -methyl- IH- indazol-5-ylamine to give the desired product as a pale pink solid (40 mg, 59%). 1H NMR (400 MHz, DMSO-40 δ ppm 0.43 - 0.49 (m, 2 H), 0.78 - 0.86 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.67 - 1.77 (m, 3 H), 1.78 - 1.90 (m, 1 H), 1.91 - 2.02 (m, 2 H), 2.05 - 2.17 (m, 2 H), 2.89 - 3.00 (m, 1 H), 3.10 - 3.21 (m, 2 H), 3.40 - 3.50 (m, 2 H), 3.98 (s, 3 H), 6.77 - 6.84 (m, 1 H), 7.47 (d, 7=8.7 Hz, 1 H), 7.57 - 7.62 (m, 2 H), 7.65 - 7.72 (m, 1 H), 7.88 - 7.92 (m, 1 H), 8.30 (s, 1 H), 8.86 (s, 1 H); m/z (ES+APCI)+: 420 [M+H]+.
Example 138
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(2-methyl-2H-indazol-6-ylamino)-pyrimidin-4- ylaminoj-propylj-amide.
Figure imgf000202_0001
Example 138 was prepared analogously to Example 94 to from Intermediate 8 and 2-methyl-2H- indazol-6-ylamine to give the desired product as a pale brown solid (39 mg, 57%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.45 - 0.51 (m, 2 H), 0.80 - 0.86 (m, 2 H), 1.43 - 1.51 (m, 1 H), 1.66 - 1.89 (m, 4 H), 1.90 - 2.00 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.89 - 3.01 (m, 1 H), 3.13 - 3.21 (m, 2 H), 3.43 - 3.52 (m, 2 H), 4.07 (s, 3 H), 6.83 - 6.89 (m, 1 H), 7.16 (dd, ./=9.2, 1.8 Hz, 1 H), 7.48 (d, 7=8.7 Hz, 1 H), 7.64 (d, 7=0.9 Hz, 1 H), 7.74 - 7.81 (m, 1 H), 8.11 - 8.14 (m, 1 H), 8.32 (s, 1 H), 8.90 (s, 1 H); m/z (ES+APCI)+: 420 [M+H]+.
Example 139
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(l-methyl-lH-indazol-6-ylamino)-pyrimidin-4- ylamino] -propyl} -amide.
Figure imgf000202_0002
Example 139 was prepared analogously to Example 94 to from Intermediate 8 and 1-methyl-lH- indazol-6-ylamine to give the desired product as an off-white solid (42 mg, 61%). 1H NMR (400
MHz, DMSO-J6) δ ppm 0.46 - 0.53 (m, 2 H), 0.82 - 0.88 (m, 2 H), 1.46 - 1.54 (m, 1 H), 1.66 - 1.90
(m, 4 H), 1.91 - 2.01 (m, 2 H), 2.03 - 2.15 (m, 2 H), 2.88 - 2.99 (m, 1 H), 3.10 - 3.19 (m, 2 H), 3.50
- 3.58 (m, 2 H), 3.93 (s, 3 H), 6.87 - 6.94 (m, 1 H), 7.19 (dd, 7=8.7, 1.8 Hz, 1 H), 7.53 (d, 7=8.7 Hz,
1 H), 7.65 - 7.71 (m, 2 H), 7.82 - 7.86 (m, 1 H), 8.42 (s, 1 H), 9.17 (s, 1 H); m/z (ES+APCI)+: 420 [M+H]+.
Example 140
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[3-(2-oxo-imidazolidin-l-yl)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide.
Figure imgf000203_0001
Example 140 was prepared analogously to Example 94 from Intermediate 8 and l-(3-amino- phenyl)-imidazolidin-2-one to give the desired product as a white solid (50 mg, 69%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.44 - 0.48 (m, 2 H), 0.80 - 0.85 (m, 2 H), 1.43 - 1.50 (m, 1 H), 1.64 - 1.77 (m, 3 H), 1.79 - 1.91 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.89 - 2.99 (m, 1 H), 3.08 - 3.14 (m, 2 H), 3.36 - 3.42 (m, 2 H), 3.42 - 3.48 (m, 2 H), 3.78 - 3.83 (m, 2 H), 6.76 - 6.81 (m, 1 H), 6.86 (br. s, 1 H), 7.08 - 7.13 (m, 1 H), 7.18 - 7.22 (m, 1 H), 7.38 - 7.41 (m, 1 H), 7.60 (d, J=0.9 Hz, 1 H), 7.64 - 7.69 (m, 1 H), 7.80 - 7.82 (m, 1 H), 8.81 (br. s, 1 H); m/z (ES+APCI)+: 450 [M+H]+.
Example 141
Cyclobutanecarboxylic acid {3-[2-(3-bromo-phenylamino)-5-cyclopropyl-pyrimidin-4-ylamino]- propylj-amide
Figure imgf000203_0002
Example 141 was prepared analogously to Example 94 to from Intermediate 8 and 3-bromo- phenylamine to give the desired product as an off-white foam (0.26 g, 92%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.45 - 0.50 (m, 2 H), 0.80 - 0.86 (m, 2 H), 1.43 - 1.51 (m, 1 H), 1.67 - 1.78 (m, 3 H), 1.79 - 1.92 (m, 1 H), 1.94 - 2.03 (m, 2 H), 2.04 - 2.17 (m, 2 H), 2.91 - 3.02 (m, 1 H), 3.09 - 3.20 (m, 2 H), 3.39 - 3.48 (m, 2 H), 6.90 - 6.95 (m, 1 H), 6.99 (ddd, ./=7.8, 1.8, 0.9 Hz, 1 H), 7.12 - 7.19 (m, 1 H), 7.57 - 7.61 (m, 1 H), 7.64 (d, J=0.9 Hz, 1 H), 7.66 - 7.71 (m, 1 H), 8.26 - 8.31 (m, 1 H), 9.14 (s, 1 H); m/z (ES+APCI)+: 444/ 446 [M+H]+.
Example 142 Cyclobutanecarboxylic acid β-fS-cyclopropyl^-β-fpiperazine-l-carbonylJ-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide. Hydrochloride.
Figure imgf000204_0001
Step l
To a solution of piperazine-1-carboxylic acid tert-butyl ester (11 mg, 0.06 mmol) in DMF (1 ml) was added Example 71 (40 mg, 0.09 mmol), HATU (36 mg, 0.09 mmol) and diisopropylethylamine (63 μl, 0.35 mmol), and the reaction was stirred at room temperature for 18 hours. The mixture was evaporated, taken up in 9:1 DCM:MeOH and filtered through a 0.5 g Isolute-NH2 cartridge, eluting with 9:1 DCM: methanol. Flash chromatography on the Biotage SP4 (0 to 10% methanol/DCM gradient), gave a colourless oil (34 mg, 100%), which was used in the next step without further purification.
Step 2 The crude product from Step 1 (50 mg, 0.09 mmol) and 4M hydrogen chloride solution in dioxan (5 ml) were combined and stirred at room temperature for 18 h and evaporated. Purification by flash chromatography using a Biotage SP4 (gradient elution from 0-20% methanol in DCM), gave the desired product as a white solid (7 mg, 22%).Η NMR (400 MHz, DMSCW6) δ ppm 0.43 - 0.50 (m, 2 H), 0.77 - 0.90 (m, 2 H), 1.42 - 1.54 (m, 1 H), 1.61 - 1.77 (m, 3 H), 1.78 - 2.03 (m, 3 H), 2.04 - 2.17 (m, 2 H), 2.65 - 2.87 (m, 4 H), 2.92 - 3.04 (m, 1 H), 3.09 - 3.15 (m, 2 H), 3.28 - 3.49 (m, 2 H), 3.51 - 3.67 (m, 4 H), 4.15 (br. s, 1 H), 6.80 - 6.85 (m, 1 H), 6.91 - 6.96 (m, 1 H), 7.23 - 7.30 (m, 1 H), 7.62 (s, 1 H), 7.74 - 7.78 (m, 1 H), 7.78 - 7.83 (m, 1 H), 7.89 - 7.93 (m, 1 H), 9.08 (br. s, 1 H): m/z (ES+APCI)+: 478 [M+H]+.
Example 143
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[3-(2-morpholin-4-yl-ethoxy)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide.
Figure imgf000205_0001
Example 143 was prepared analogously to Example 64 from Intermediate 8 and 3-(2-morpholin-4- yl-ethoxy)-phenylamine to give the desired product as a white foam (15 mg, 23%).Η NMR (400 MHz, DMSO-c4) δ ppm 0.44 - 0.49 (m, 2 H), 0.80 - 0.86 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.64 - 1.77 (m, 3 H), 1.79 - 1.92 (m, 1 H), 1.93 - 2.02 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.44 - 2.48 (m, 4 H), 2.67 (t, /=5.7 Hz, 2 H), 2.90 - 3.00 (m, 1 H), 3.10 - 3.17 (m, 2 H), 3.40 - 3.47 (m, 2 H), 3.53 - 3.60 (m, 4 H), 4.02 (t, 7=5.7 Hz, 2 H), 6.39 - 6.46 (m, 1 H), 6.82 - 6.90 (m, 1 H), 7.03 - 7.12 (m, 1 H), 7.21 - 7.25 (m, 1 H), 7.57 - 7.59 (m, 1 H), 7.61 (d, 7=0.9 Hz, 1 H), 7.65 - 7.72 (m, 1 H), 8.87 (br. s, 1 H): m/z (ES+APCI)+: 495 [M+H]+.
Example 144
Pyrrolidine- 1-carboxylic acid (3-{4-[3-(cyclobutanecarbonyl-amino)-propylamino]-5-cyclopropyl- pyrimidin-2-ylamino}-phenyl)-amide.
Figure imgf000205_0002
Example 144 was prepared analogously to Example 64 from Intermediate 8 and Intermediate 10 to give the desired product as a white foam (5 mg, 8%). 1H NMR (400 MHz, DMSO^6) δ ppm 0.42 - 0.48 (m, 2 H), 0.79 - 0.85 (m, 2 H), 1.42 - 1.50 (m, 1 H), 1.63 - 1.77 (m, 3 H), 1.80 - 1.91 (m, 5 H), 1.92 - 2.01 (m, 2 H), 2.04 - 2.15 (m, 2 H), 2.89 - 2.99 (m, 1 H), 3.08 - 3.14 (m, 2 H), 3.32 - 3.38 (m, 4 H), 3.42 - 3.48 (m, 2 H), 6.73 - 6.78 (m, 1 H), 6.90 - 6.94 (m, 1 H), 7.00 - 7.06 (m, 1 H), 7.27 - 7.30 (m, 1 H), 7.58 (d, /=0.9 Hz, 1 H), 7.64 - 7.69 (m, 1 H), 7.87 - 7.90 (m, 1 H), 7.94 (br. s, 1 H), 8.73 (br. s, 1 H); m/z (ES+APCI)+: 478 [M+H]+.
Example 145 Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[3-(l-methyl-l-morpholin-4-yl-ethyl)- phenylamino]-pyrimidin-4-ylamino} -propyl) -amide
Figure imgf000206_0001
Prepared analogously to Example 91 using Intermediate 8 and 3-(l-methyl-l-morpholin-4-yl- ethyl)-phenylamine to provide the product as a white solid (24 mg, 38%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.43 - 0.48 (m, 2 H), 0.79 - 0.85 (m, 2 H), 1.27 (s, 6 H), 1.42 - 1.49 (m, 1 H), 1.64 - 1.76 (m, 3 H), 1.80 - 1.88 (m, 1 H), 1.92 - 2.01 (m, 2 H), 2.04 - 2.15 (m, 2 H), 2.31 - 2.40 (m, 4 H), 2.90 - 2.97 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.43 - 3.51 (m, 2 H), 3.51 - 3.57 (m, 4 H), 6.79 (t, 7=5.95 Hz, 1 H), 6.99 (d, 7=8.24 Hz, 1 H), 7.13 (t, 7=8.01 Hz, 1 H), 7.59 (s, 1 H), 7.61 - 7.70 (m, 2 H), 7.85 (s, 1 H), 8.80 (s, 1 H); m/z (ES+APCI)+: 493 [M+H]+.
Example 146
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-((S)-3-dimethylamino-pyrrolidin-l-ylmethyl)- pyridin-3-ylamino]-pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000206_0002
A solution of (S)-(-)-3-dimethylaminopyrrolidine (14.4 μl, 0.114 mmol) and DIPEA (79 μl, 0.455 mmol) in DCM (1 ml) was added to a stirred suspension of Intermediate 22 in DCM (1 ml), and the resultant solution was stirred at rt for 3 days. The mixture was concentrated to dryness and the resulting residue was dissolved in 10:1 DCM-MeOH, and the resulting solution was passed through a plug of silica gel. The filtrate was concentrated to dryness and the residue was purified by mass- triggered preparative HPLC (low pH buffer). The purified material was passed through a SCX cartridge to provide the product as the free base (1.4 mg). 1H NMR (400 MHz, CHLOROFORM-c/) δ ppm 0.49 - 0.54 (m, 2 H), 0.91 - 0.97 (m, 2 H), 1.45 - 1.52 (m, 1 H), 1.61 - 2.05 (m, 8 H), 2.07 - 2.16 (m, 2 H), 2.20 - 2.42 (m, 7 H), 2.54 - 2.61 (m, 1 H), 2.76 - 2.85 (m, 2 H), 2.88 - 3.02 (m, 2 H), 3.37 (q, 7=6.41 Hz, 2 H), 3.56 (q, 7=6.26 Hz, 2 H), 3.69 - 3.79 (m, 2 H), 5.87 (t, 7=6.18 Hz, 1 H), 6.04 (t, 7=6.18 Hz, 1 H), 6.90 (br. s, 1 H), 7.31 (d, 7=7.56 Hz, 1 H), 7.73 (s, 1 H), 7.96 (dd, 7=8.47,
2.52 Hz, 1 H), 8.84 (d, 7=2.29 Hz, 1 H); m/z (ES+APCI)+ : 493 [M+H]+.
Example 147 Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(4-methyl-piperazin-l-ylmethyl)-pyridin-3- ylamino]-pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000207_0001
Prepared analogously to Example 146 using Intermediate 22 and N-methylpiperazine to provide the product as the free base (1.4 mg). 1H NMR (400 MHz, CHLOROFORM-cf) δ ppm 0.49 - 0.54 (m, 2 H), 0.88 - 0.97 (m, 2 H), 1.45 - 1.60 (m, 1 H), 1.64 - 2.00 (m, 4 H), 2.07 - 2.17 (m, 2 H), 2.20 - 2.41 (m, 5 H), 2.41 - 2.71 (m, 8 H), 2.93 - 3.00 (m, 1 H), 3.38 (q, 7=6.41 Hz, 2 H), 3.53 - 3.64 (m, 4 H), 5.85 (t, 7=6.18 Hz, 1 H), 6.06 (t, 7=5.95 Hz, 1 H), 6.93 (br. s, 1 H), 7.31 (m, IH), 7.72 (m, 1 H), 7.97 (dd, 7=8.47, 2.52 Hz, 1 H), 8.85 (m, 1 H); m/z (ES+APCI)+ : 479 [M+H]+.
Example 148
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(4,4-difluoro-piperidin-l-ylmethyl)-pyridin-3- ylamino]-pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000207_0002
Prepared analogously to Example 146 using Intermediate 22 and 4,4-difluoropiperidine HCl to provide the product as the free base (14 mg). 1H NMR (400 MHz, CHLOROFORM-α!) δ ppm 0.49 - 0.57 (m, 2 H), 0.88 - 0.99 (m, 2 H), 1.45 - 1.54 (m, 1 H), 1.75 - 2.17 (m, 10 H), 2.19 - 2.33 (m, 2 H), 2.57 - 2.65 (m, 4 H), 2.97 (quin, 7=8.70 Hz, 1 H), 3.38 (q, 7=6.11 Hz, 2 H), 3.48 - 3.70 (m, 4 H), 5.79 (br. s, 1 H), 6.11 (t, 7=5.95 Hz, 1 H), 7.05 - 7.14 (m, 1 H), 7.29 (d, 7=8.24 Hz, 1 H), 7.71 (s, 1 H), 8.01 (dd, 7=8.47, 2.52 Hz, 1 H), 8.81 - 8.86 (m, 1 H), m/z (ES+APCI)+ : 500 [M+H]+.
Example 149 Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(pyrimidin-5-ylamino)-pyrimidin-4-ylamino]- propyl} -amide
Figure imgf000208_0001
Intermediate 8 (40 mg, 0.130 mmol), 5-aminopyrimidine (14.8 mg, 0.156 mmol), xantphos (6.0 mg, 0.010 mmol), sodium t-butoxide (37 mg, 0.390 mmol) and Pd2(dba)3 (7.1 mg, 0.008 mmol) were placed in a sealed tube. The mixture was degassed, placed under an atmosphere of nitrogen and heated at 1000C overnight. The mixture was diluted with EtOAc and water. The organic phase was washed with brine, dried and concentrated. The residue was purified by mass-triggered preparative HPLC (low pH buffer) to afford a colourless oil. This material was passed through a SCX cartridge to provide the product as the free base (2.4 mg, 5 %). 1H NMR (400 MHz, DMSCW6) δ ppm 0.46 - 0.54 (m, 2 H), 0.80 - 0.90 (m, 2 H), 1.45 - 1.52 (m, 1 H), 1.65 - 1.77 (m, 3 H), 1.81 - 1.90 (m, 1 H), 1.93 - 2.16 (m, 4 H), 2.97 (quin, 7=8.24 Hz, 1 H), 3.09- 3.16 (m, 2 H), 3.35 - 3.46 (m, 2 H), 6.99 (t, /=5.72 Hz, 1 H), 7.63 - 7.71 (m, 2 H), 8.67 (s, 1 H), 9.15 (s, 2 H), 9.29 (s, 1 H); m/z (ES+APCI)+ : 368 [M+H]+.
Example 150
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(3, 4, 5, 6-tetrahydro-2H-[l ,2J ' bipyridinyl-5'- ylamino)-pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000208_0002
Prepared analogously to Example 97 using Intermediate 8 and 3,4,5,6-tetrahydro-2H- [l,2']bipyridinyl-5'-ylamine to provide product as the free base (31 mg, 53 %). 1H NMR (400 MHz, DMSCW6) δ ppm 0.40 - 0.45 (m, 2 H), 0.76 - 0.84 (m, 2 H), 1.39 - 1.47 (m, 1 H), 1.48 - 1.59 (m, 6 H), 1.62 - 1.76 (m, 3 H), 1.80 - 1.90 (m, 1 H), 1.92 - 2.01 (m, 2 H), 2.04 - 2.15 (m, 2 H), 2.90 - 3.00 (m, 1 H), 3.07 - 3.17 (m, 2 H), 3.33 - 3.42 (m, 6 H), 6.71 - 6.78 (m, 2 H), 7.53 (s, 1 H), 7.65 (t, 7=5.72 Hz, 1 H), 7.89 (dd, ./=9.16, 2.75 Hz, 1 H), 8.37 (d, 7=2.75 Hz, 1 H), 8.58 (s, 1 H); m/z
(ES+APCI)+ : 450 [M+H]+.
Example 151 Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-pyrrolidin-l-yl-pyridin-3-ylaτnino)-pyrimidin- 4-ylamino]-propyl}-amide
Figure imgf000209_0001
Prepared analogously to Example 97 using Intermediate 8 and 6-pyrrolidin-l-yl-pyridin-3-ylamine to provide product as the free base (31 mg, 55 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.39 - 0.46 (m, 2 H), 0.77 - 0.85 (m, 2 H), 1.40 - 1.47 (m, 1 H), 1.62 - 1.78 (m, 3 H), 1.81 - 2.02 (m, 7 H), 2.05 - 2.16 (m, 2 H), 2.96 (quin, 7=8.24 Hz, 1 H), 3.07 - 3.18 (m, 2 H), 3.26 - 3.43 (m, 6 H), 6.38 (d, /=9.16 Hz, 1 H), 6.75 (t, 7=5.72 Hz, 1 H), 7.53 (s, 1 H), 7.66 (t, 7=5.95 Hz, 1 H), 7.84 (dd, 7=8.70, 2.75 Hz, 1 H), 8.31 (d, 7=2.29 Hz, 1 H)1 8.48 (s, 1 H); m/z (ES+ APC I)+ : 436 [M+H]+.
Example 152
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(2-morpholin-4-yl-pyrimidin-5-ylamino)- pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000209_0002
Prepared and analogously to Example 149 using Intermediate 8 and Intermediate 47 to give a yellow solid (9 mg, 15 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.41 - 0.48 (m, 2 H), 0.78 - 0.86 (m, 2 H), 1.40 - 1.48 (m, 1 H), 1.61 - 1.77 (m, 3 H), 1.81 - 1.91 (m, 1 H), 1.92 - 2.16 (m, 4 H), 2.91 - 3.02 (m, 1 H), 3.06 - 3.17 (m, 2 H), 3.29 - 3.42 (m, 2 H), 3.55 - 3.69 (m, 8 H), 6.86 (br. s, 1 H), 7.53 - 7.57 (m, 1 H), 7.66 (d, 7=5.95 Hz, 1 H), 8.63 - 8.74 (m, 3 H); m/z (ES+APCI)+ : 453 [M+H]+.
Example 153 Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(l-methyl-lH-pyrazol-4-ylamino)-pyrimidin-4- ylaminoj-propylj-amide
Figure imgf000210_0001
Prepared and analogously to Example 149 using Intermediate 8 and 1 -methyl- lH-pyrazol-4- ylamine to provide a white solid (17 mg, 35 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.40 - 0.47 (m, 2 H), 0.76 - 0.85 (m, 2 H), 1.39 - 1.47 (m, 1 H), 1.60 - 1.78 (m, 3 H), 1.82 - 1.92 (m, 1 H), 1.94
- 2.03 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.97 (quin, /=8.36 Hz, 1 H), 3.06 - 3.16 (m, 2 H), 3.40 (q, 7=6.41 Hz, 2 H), 3.76 (s, 3 H), 6.71 (t, 7=5.72 Hz, 1 H), 7.40 (s, 1 H), 7.54 (s, 1 H), 7.69 (t, /=5.14 Hz, 1 H), 7.76 (s, 1 H), 8.65 (br. s, 1 H); m/z (ES+ APCI)+ : 370 [M+H]+.
Example 154
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(2-morpholin-4-yl-ethoxy)-pyridin-3-ylamino]- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000210_0002
Prepared analogously to Example 97 using Intermediate 8 and Intermediate 49 to give an off-white solid (20 mg, 25 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.42 - 0.50 (m, 2 H), 0.79 - 0.87 (m, 2 H), 1.41 - 1.48 (m, 1 H), 1.63 - 1.77 (m, 3 H), 1.81 - 1.89 (m, 1 H), 1.93 - 2.02 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.45 (br. s, 4 H), 2.66 (t, /=5.72 Hz, 2 H), 2.92 - 2.99 (m, 1 H), 3.08 - 3.15 (m, 2 H), 3.35
- 3.42 (m, 2 H), 3.53 - 3.59 (m, 4 H), 4.27 - 4.34 (m, 2 H), 6.72 (d, /=9.16 Hz, 1 H), 6.82 (t, /=5.95 Hz, 1 H), 7.58 (s, 1 H), 7.64 - 7.69 (m, 1 H), 8.04 (dd, /=8.70, 2.75 Hz, 1 H), 8.44 (d, /=2.29 Hz, 1 H), 8.80 (s, 1 H); m/z (ES+APCI)+ : 496 [M+H]+.
Example 155
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[l-(2-morpholin-4-yl-ethyl)-lH-pyrazol-4- ylamino]-pyrimidin-4-ylamino}-propyl) -amide
Figure imgf000211_0001
Prepared and analogously to Example 149 using Intermediate 8 and
Intermediate 51 to give the product as an off-white coloured solid (17 mg, 28 %). 1H NMR (400
MHz, DMS(W6) δ ppm 0.41 - 0.47 (m, 2 H), 0.78 - 0.85 (m, 2 H), 1.40 - 1.47 (m, 1 H), 1.61 - 1.78 (m, 3 H), 1.82 - 1.90 (m, 1 H), 1.94 - 2.03 (m, 2 H), 2.06 - 2.16 (m, 2 H), 2.33 - 2.44 (m, 4 H), 2.64 - 2.72 (m, 2 H), 2.93 - 3.00 (m, 1 H), 3.06 - 3.16 (m, 2 H), 3.35 - 3.45 (m, 2 H), 3.51 - 3.57 (m, 4 H), 4.12 - 4.19 (m, 2 H), 6.71 (t, 7=5.95 Hz, 1 H), 7.43 (s, 1 H), 7.55 (s, 1 H), 7.65 - 7.72 (m, 1 H), 7.82 (s, 1 H), 8.66 (br. s, 1 H),. m/z (ES+ APCI)+ : 469 M+H]+.
Example 156
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-piperidin-l-ylmethyl-pyridin-3-ylamino)- pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000211_0002
Prepared analogously to Example 146 using Intermediate 22 and piperidine to provide the product as the free base (7.5 mg). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.44 - 0.49 (m, 2 H), 0.80 - 0.86 (m, 2 H), 1.34 - 1.53 (m, 7 H), 1.65 - 1.77 (m, 3 H), 1.81 - 1.89 (m, 1 H), 1.93 - 2.02 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.34 (br. s, 4 H), 2.93 - 3.00 (m, 1 H), 3.09 - 3.16 (m, 2 H), 3.36 - 3.49 (m, 4 H), 6.87 (t, 7=5.95 Hz, 1 H), 7.25 (d, 7=8.24 Hz, 1 H), 7.61 (s, 1 H), 7.69 (t, 7=5.72 Hz, 1 H), 8.16 (dd, 7=8.47, 2.52 Hz, 1 H), 8.77 (d, 7=2.29 Hz, 1 H), 9.04 (s, 1 H); m/z (ES+APCI)+ : 464 [M+H]+.
Example 157
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(3l3-difluoro-piperidin-]-ylmethyl)-pyridin-3- ylamino]-pyrimidin-4-ylamino}-propyl) -amide
Figure imgf000212_0001
Prepared analogously to Examp-le 146 using Intermediate 22 and 3,3-difluoropiperidine HCl to provide the product as the free base (16 mg). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.44 - 0.49 (m, 2 H), 0.80 - 0.86 (m, 2 H), 1.43 - 1.51 (m, 1 H), 1.60 - 1.77 (m, 5 H), 1.79 - 2.02 (m, 5 H), 2.05 - 2.16 (m, 2 H), 2.42 (t, 7=4.81 Hz, 2 H), 2.61 - 2.69 (m, 2 H), 2.92 - 3.00 (m, 1 H), 3.09 - 3.17 (m, 2 H), 3.42 (q, /=6.41 Hz, 2 H), 3.59 (s, 2 H), 6.88 (t, 7=5.72 Hz, 1 H), 7.25 (d, 7=8.24 Hz, 1 H), 7.62 (s, 1 H), 7.69 (t, 7=5.72 Hz, 1 H), 8.19 (dd, 7=8.70, 2.75 Hz, 1 H), 8.80 (d, 7=2.75 Hz, 1 H), 9.08 (s, 1 H); m/z (ES+APCI)+ : 500 [M+H]+.
Example 158
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-(2-morpholin-4-yl-ethylamino)-pyridin-3- ylaminoJ-pyrimidin-4-ylamino} -propyl) -amide
Figure imgf000212_0002
Prepared analogously to Example 97 using Intermediate 8 and N2-(2-morpholin-4-yl-ethyl)- pyridine-2,5-diamine to give an off-white coloured solid (20 mg, 31 %). 1H NMR (400 MHz, DMSO-40 δ ppm 0.39 - 0.44 (m, 2 H), 0.76 - 0.82 (m, 2 H), 1.38 - 1.46 (m, 1 H), 1.61 - 1.77 (m, 3 H), 1.81 - 1.91 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.05 - 2.15 (m, 2 H), 2.36 - 2.48 (m, 6 H), 2.95 (quin, 7=8.24 Hz, 1 H), 3.06 - 3.14 (m, 2 H), 3.25 - 3.31 (m, 2 H), 3.35 - 3.41 (m, 2 H), 3.53 - 3.60 (m, 4 H), 5.88 (t, 7=5.72 Hz, 1 H), 6.41 (d, 7=8.70 Hz, 1 H), 6.71 (t, 7=5.72 Hz, 1 H), 7.51 (s, 1 H), 7.63 - 7.73 (m, 2 H), 8.21 (d, 7=2.75 Hz, 1 H), 8.40 (s, 1 H); m/z (ES+APCI)+ : 495 [M+H]+.
Example 159
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[6-((R)-3-diτnethylamino-pyrrolidin-l -ylmethyl)- pyridin-3-ylamino]-pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000213_0001
Prepared analogously to Example 146 using Intermediate 22 and (R)-(+)-3- dimethylaminopyrrolidine to provide the product as the free base (8 mg). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.44 - 0.49 (m, 2 H), 0.80 - 0.86 (m, 2 H), 1.43 - 1.50 (m, 1 H), 1.54 - 1.63 (m, 1 H), 1.65 - 1.91 (m, 5 H), 1.93 - 2.16 (m, 10 H), 2.22 - 2.30 (m, 1 H), 2.44 (td, /=8.82, 6.18 Hz, 1 H), 2.52 - 2.71 (m, 3 H), 2.97 (quin, /=8.24 Hz, 1 H), 3.13 (q, 7=6.41 Hz, 2 H), 3.37 - 3.47 (m, 2 H), 3.52 (d, 7=13.28 Hz, 1 H), 3.61 (d, /=13.28 Hz, 1 H), 6.88 (t, /=5.95 Hz, 1 H), 7.23 (d, /=8.70 Hz, 1 H), 7.61 (s, 1 H), 7.69 (t, /=5.72 Hz, 1 H), 8.14 - 8.18 (m, 1 H), 8.77 (d, /=2.75 Hz, 1 H), 9.04 (s, 1 H); m/z (ES+APCI)+ : 493 [M+H]+.
Example 160
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[l-(2-piperidin-l-yl-ethyl)-lH-pyrazol-4- ylamino]-pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000213_0002
Prepared and analogously to Example 149 using Intermediate 8 and Intermediate 53 to give the product as the free base (19 mg, 31 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.38 - 0.47 (m, 2 H), 0.75 - 0.85 (m, 2 H), 1.29 - 1.49 (m, 7 H), 1.60 - 1.77 (m, 3 H), 1.79 - 1.91 (m, 1 H), 1.92 - 2.02 (m, 2 H), 2.04 - 2.15 (m, 2 H), 2.34 (br. s, 4 H), 2.63 (t, /=6.87 Hz, 2 H), 2.96 (quin, /=8.24 Hz, 1 H), 3.04 - 3.16 (m, 2 H), 3.40 (q, /=6.1 1 Hz, 2 H), 4.10 (t, /=6.87 Hz, 2 H), 6.70 (t, /=5.50 Hz, 1 H), 7.41 (s, 1 H), 7.53 (s, 1 H), 7.69 (t, /=5.72 Hz, 1 H), 7.80 (s, 1 H), 8.65 (br. s, 1 H); m/z (ES+APCI)+ : 467 [M+H]+.
Example 161
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(l-isopropyl-lH-pyrazol-4-ylamino)-pyrimidin-4- ylamino] -propyl} -amide
Figure imgf000214_0001
Prepared and analogously to Example 149 using Intermediate 8 and l-isopropyl-lH-pyrazol-4- ylamine to give the product as a white foamy solid (22 mg, 34 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.36 - 0.45 (m, 2 H), 0.75 - 0.84 (m, 2 H), 1.32 - 1.47 (m, 7 H), 1.60 - 1.77 (m, 3 H), 1.79 - 1.91 (m, 1 H), 1.93 - 2.02 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.96 (quin, /=8.24 Hz, 1 H), 3.05 - 3.17 (m, 2 H), 3.35 - 3.47 (m, 2 H), 4.39 (quin, J=6.64 Hz, 1 H), 6.71 (t, J=5.50 Hz, 1 H), 7.41 (s, 1 H), 7.54 (s, 1 H), 7.68 (t, J=5.72 Hz, 1 H), 7.82 (s, 1 H), 8.65 (br. s, 1 H); m/z (ES+APCI)+ : 398 [M+H]+.
Example 162
Cyclobutyl-(3-{[5-cyclopropyl-2-(3-fluoro-phenylamino)-pyrimidin-4-ylcmιino]-inethyl}-azetidin-l- yl)-methanone
Figure imgf000214_0002
A solution of Intermediate 56 (37 mg, 0.1 mmol), 3-fluoroaniline (50 mg, 0.46 mmol) and glacial AcOH (3 drops) in n-butanol (2 ml) was heated at 150 0C in the microwave for 35 min. The crude mixture was concentrated under vacuum and purified by LCMS (low pH buffer). 1H NMR (400
MHz, DMS(W6) δ ppm 0.48 - 0.65 (m, 2 H), 0.80 - 0.97 (m, 2 H), 1.60 - 1.80 (m, 2 H), 1.81 - 2.29
(m, 6 H), 2.85 - 3.08 (m, 1 H), 3.11 - 3.57 (m, 3 H), 3.57 - 3.75 (m, 2 H), 3.76 - 3.92 (m, 1 H), 4.47
- 4.72 (m, 1 H), 6.83 - 7.01 (m, 1 H), 7.20 - 7.44 (m, 2 H), 7.51 - 7.70 (m, 2 H); m/z (ES+APCI)+ : 396 [M+H]+. Example 163
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[4-(pyrimidin-2-ylsulfamoyl)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000215_0001
A solution of Intermediate 8 (50 mg, 0.16 mmol), 4-amino-N-pyrimidin-2-yl-benzenesulfonamide (162 mg, 0.65 mmol) and glacial AcOH (37 μl) in n-Butanol was heated at 150 0C in the microwave for 35 min. The precipitate was filtered and the filtrate was concentrated under vacuum and purified by preparative LCMS (low pH buffer). The resulting product was run through an amino propyl cartridge to give the free base (9.4 mg, 11%). 1H NMR (400 MHz, CHLOROFORM- d) δ ppm 0.46 - 0.63 (m, 2 H), 1.00 (d, 7=6.87 Hz, 2 H), 1.54 (d, /=4.12 Hz, 1 H), 1.68 - 1.80 (m, 2 H), 1.80 - 1.89 (m, 1 H), 1.90 - 2.00 (m, 1 H), 2.07 - 2.19 (m, 2 H), 2.19 - 2.32 (m, 2 H), 2.63 (s, 1 H), 2.91 - 3.11 (m, 1 H), 3.36 (q, /=5.95 Hz, 2 H), 3.52 - 3.63 (m, 2 H), 5.87 (t, 7=6.41 Hz, 1 H), 6.83 (br. s, 1 H), 6.94 (t, 7=4.81 Hz, 1 H), 7.77 (d, 7=9.16 Hz, 3 H), 8.01 (d, 7=8.70 Hz, 2 H), 8.59 (d, 7=4.58 Hz, 2 H), 8.73 (br. s, 1 H); m/z (ES+APCI)+ : 523 [M+H]+.
Examples 164-170
Examples 164-170 were prepared analogously to Example 163 (the general structure is shown below followed by the tabulated examples).
Figure imgf000216_0001
Figure imgf000216_0002
Figure imgf000217_0002
a: HPLC column: 21.2xl00mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 30ml/min; Run time: 12.0 min: Solvent A: 0.1% Trifluoro acetic acid in water, Solvent B: Methanol; Gradient - 10-100%B; Gradient time: 9.0 min. b: HPLC column: 4.6x50mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 2ml/min; Run time: 4.6 min: Solvent A: 0.1% Ammonium Hydroxide in water, Solvent B: Methanol; Gradient - 10- 100%B; Gradient time: 3.5min.
Example 171
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[4-(l-methyl-piperidin-4-yl)-phenylaminoJ- pyriτnidin-4-ylamino}-propyl)-amide
Figure imgf000217_0001
A solution of Intermediate 8 (45 mg, 0.15 mmol), 4-(l-methyl-piperidin-4-yl)-phenylamine (33 mg, 0.18 mmol), Pd2(dba)3 (8 mg, 0.009 mmol), xantphos (7 mg, 0.012 mmol) and NaO1Bu (42 mg, 0.45 mmol) in dioxane was degassed and heated at 90 0C under N2 overnight. The reaction mixture was concentrated under vacuum and purified by silica gel chromatography followed by preparative
LCMS, to give the desired product (14 mg, 20%). 1H NMR (400 MHz, CHLOROFORM-cO δ ppm 0.44 - 0.58 (m, 2 H), 0.85 - 0.95 (m, 2 H), 1.39 - 1.54 (m, 1 H), 1.69 - 1.95 (m, 7 H), 1.97 - 2.11 (m, 4 H), 2.13 - 2.29 (m, 2 H), 2.33 (s, 3 H), 2.37 - 2.50 (m, 1 H), 2.67 - 2.82 (m, 1 H), 2.97 (d, 7=10.53 Hz, 2 H), 3.32 (q, 7=6.41 Hz, 2 H), 3.48 (s, 1 H), 3.60 (q, 7=5.95 Hz, 2 H), 5.76 (t, 7=6.18 Hz, 1 H), 5.82 (t, 7=6.18 Hz, 1 H), 6.90 (s, 1 H), 7.16 (d, 7=8.70 Hz, 2 H), 7.52 (d, 7=8.70 Hz, 2 H), 7.72 (s, 1 H); m/z (ES+APCI)+ : 463 [M+H]+.
Example 172
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[4-(2-dimethylamino-ethyl)-phenylaminoJ- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000218_0001
A solution of Intermediate 8 (45 mg, 0.15 mmol), 4-(2-dimethylamino-ethyl)-phenylamine (30 mg, 0.18 mmol), Pd2(dba)3 (8 mg, 0.009 mmol), xantphos (7 mg, 0.012 mmol) and NaO1Bu (42 mg, 0.45 mmol) in dioxane was degassed and heated at 90 0C under N2 overnight. The reaction mixture was purified silica gel chromatography followed by preparative LCMS. The resulting residues were run through an Isolute-NH2 cartridge, eluting with MeOH, and concentrated under vacuum to give the desired product (10.2 mg, 16 %). 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.45 - 0.55 (m, 2 H), 0.85 - 0.97 (m, 2 H), 1.39 - 1.53 (m, 1 H), 1.68 - 1.96 (m, 4 H), 1.96 - 2.10 (m, 2 H), 2.15 - 2.28 (m, 2 H), 2.30 (s, 6 H), 2.47 - 2.56 (m, 2 H), 2.68 - 2.78 (m, 3 H), 3.31 (q, 7=6.41 Hz, 2 H), 3.60 (q, 7=6.41 Hz, 2 H), 5.79 (q, 7=6.26 Hz, 2 H), 7.00 (s, 1 H), 7.14 (d, 7=8.70 Hz, 2 H), 7.51 (d, 7=8.24 Hz, 2 H), 7.73 (s, 1 H); m/z (ES+APCI)+ : 437 [M+H]+.
Example 173 Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[4-(2-dimethylamino-methyl)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000219_0001
Prepared analogously to Example 172 from Intermediate 8 (45 mg, 0.15 mmol) to give the product (9.7 mg, 15%). 1H NMR (400 MHz, CHLOROFORM-rf) δ ppm 0.46 - 0.53 (m, 2 H), 0.86 - 0.96 (m, 2 H), 1.40 - 1.52 (m, 1 H), 1.71 - 1.96 (m, 4 H), 1.99 - 2.12 (m, 2 H), 2.17 - 2.30 (m, 8 H), 2.78 (quin, 7=8.36 Hz, 1 H), 3.24 - 3.36 (m, 2 H), 3.37 (s, 2 H), 3.59 (q, 7=6.41 Hz, 2 H), 5.78 (t, J=6.18 Hz, 1 H), 5.87 (t, /=6.41 Hz, 1 H), 7.08 (s, 1 H), 7.22 (d, 7=8.70 Hz, 2 H), 7.56 (d, 7=8.70 Hz, 2 H), 7.73 (s, 1 H); m/z (ES+APCI)+ : 423 [M+H]+.
Example 174
Cyclobutanecarboxylic acid β-fS-cyclopropyl^-ft-fmorpholine^-carbonylJ-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide
Figure imgf000219_0002
A solution of Intermediate 57 (40 mg, 0.1 mmol), morpholine (9 mg, 0.1 mmol), DIPEA (29 μl, 0.3 mmol) and HATU (42 mg, 0.11 mmol) in DMF was stirred at RT for 18 h. The reaction was concentrated under vacuum and purified by preparative LCMS (low pH buffer). The resulting fractions were combined and run through a carbonate cartridge, eluting with MeOH, then concentrated under vacuum to give the product (4.4 mg, 9 %). 1H NMR (400 MHz, DMSO-d6) δ ppm 0.43 - 0.51 (m, 2 H), 0.78 - 0.87 (m, 2 H), 1.41 - 1.53 (m, 1 H), 1.65 - 1.78 (m, 3 H), 1.80 - 1.92 (m, 1 H), 1.93 - 2.03 (m, 2 H), 2.03 - 2.18 (m, 2 H), 2.97 (t, J=8.47 Hz, 1 H), 3.09 - 3.19 (m, 2 H), 3.43 (q, J=6.41 Hz, 3 H), 3.50 (br. s, 3 H), 3.55 - 3.65 (m, 4 H), 6.89 (t, J=5.72 Hz, 1 H), 7.31 (d, J=8.70 Hz, 2 H), 7.63 (s, 1 H), 7.70 (t, J=5.72 Hz, 1 H), 7.83 (d, J=8.70 Hz, 2 H), 9.17 (s, 1 H); m/z (ES+APCI)+ : 479 [M+H]+.
Examples 175-176
Prepared analogously to Example 174 (the general structure is shown below followed by the tabulated examples).
Figure imgf000220_0001
Figure imgf000220_0002
Figure imgf000221_0002
*HPLC column: 21.2xl00mm (5μm) C-18 Phenomenex Gemini; flow rate: 20ml/min; run time: 10 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: 0.1% trifluoroacetic acid is added to the water.
Example 177
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[4-(2-hydroxy-ethyl)-phenylamino]-pyrimidin-4- ylamino} -propyl) -amide
Figure imgf000221_0001
A solution of Intermediate 8 (40 mg, 0.13 mmol), 2-(4-amino-phenyl)-ethanol (55 mg, 0.4 mmol) and glacial AcOH (2 mg) in H-butanol heated at 150 0C in the microwave for 30 min. The resulting precipitate was filtered and washed with MeOH followed by petroleum ether to give the desired product (16 mg, 30 %). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.51 - 0.60 (m, 2 H), 0.87 - 0.94 (m, 2 H), 1.48 - 1.59 (m, 1 H), 1.67 - 1.78 (m, 3 H), 1.79 - 1.92 (m, 1 H), 1.93 - 2.02 (m, 2 H), 2.04 - 2.16 (m, 2 H), 2.71 (t, 7=7.10 Hz, 2 H), 2.95 (t, 7=8.47 Hz, 1 H), 3.07 - 3.17 (m, 2 H), 3.46 (q, 7=6.41 Hz, 2 H), 3.60 (t, 7=7.10 Hz, 2 H), 4.65 (br. s, 1 H), 7.24 (d, 7=8.70 Hz, 2 H), 7.45 (d, 7=8.24 Hz, 2 H), 7.56 (s, 1 H), 7.76 (t, 7=5.72 Hz, 1 H), 8.56 (br. s, 1 H), 10.18 (s, 1 H); m/z (ES+APCI)+ : 409 [M+H]+.
Example 178
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(2-oxo-2,3-dihydro-lH-benzimidazol-5-ylamino)- pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000222_0001
A solution of Intermediate 8 (80 mg 0.26 mmol), 5-amino-l,3-dihydro-benzimidazol-2-one (77 mg, 0.52 mmol) and glacial AcOH (cat.) in n-butanol was heated at 150 0C in the microwave for 35 min. The precipitate was removed and the filtrate was concentrated under vacuum and triturated in EtOAc yielding a second precipitate. This was purified by preparative LCMS (low pH buffer) to give the product (11 mg, 10 %). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.52 - 0.60 (m, 2 H), 1.47 - 1.58 (m, 1 H), 1.67 - 1.78 (m, 3 H), 1.79 - 1.93 (m, 1 H), 1.93 - 2.03 (m, 2 H), 2.03 - 2.16 (m, 2 H), 2.88 - 3.02 (m, 1 H), 3.05 - 3.15 (m, 2 H), 3.45 (q, J=6.41 Hz, 2 H), 6.90 - 7.02 (m, 2 H), 7.24 (br. s, 1 H), 7.47 (br. s, 1 H), 7.75 (t, 7=5.72 Hz, 1 H), 8.48 (t, J=5.5O Hz, 1 H), 10.16 (s, 1 H), 10.66 (s, 1 H), 10.77 (s, 1 H); m/z (ES+APCI)+ : 422 [M+H]+.
Example 179
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(l,3-dimethyl-2-oxo-2, 3-dihydro-lH- benzimidazol-5-ylamino)-pyrimidin-4-ylamino] -propyl} -amide
Figure imgf000222_0002
Prepared analogously to Example 178 from Intermediate 8 and 5-amino-l,3-dimethyl-l,3-dihydro- 2H-benzimidazol-2-one to give the desired product (32 mg, 27 %). 1H NMR (400 MHz, DMSO-ύT6) δ ppm 0.51 - 0.60 (m, 2 H), 0.88 - 0.94 (m, 2 H), 1.13 - 1.20 (m, 4 H), 1.50 - 1.59 (m, 1 H), 1.66 -
1.77 (m, 3 H), 1.83 - 1.89 (m, 1 H), 1.93 - 2.01 (m, 4 H), 2.02 - 2.13 (m, 2 H), 2.93 (t, ./=8.24 Hz, 1 H), 3.04 - 3.13 (m, 2 H), 3.47 (q, 7=6.26 Hz, 2 H), 4.03 (q, 7=6.87 Hz, 2 H), 7.06 - 7.13 (m, 1 H), 7.15 - 7.20 (m, 1 H), 7.45 (s, 1 H), 7.51 (br. s, 1 H), 7.73 (t, 7=5.50 Hz, 1 H); m/z (ES+APCI)+ : 450 [M+H]+.
Example 180
4-{4-[3-(Cyclobutanecarbonyl-amino)-propylamino]-5-cyclopropyl-pyrimidin-2-ylamirιo}-N,N- dimethyl-benzam ide
Figure imgf000223_0001
A solution of Intermediate 8 (50 mg, 0.16 mmol), 4-amino-N,N-dimethyl-benzamide (78 mg, 0.49 mmol) and glacial AcOH (cat.) in w-butanol was heated at 1500C in the microwave for 35 min. The crude reaction mixture was concentrated under vacuum and purified by prep LCMS (high pH buffer) to give the product (40 mg, 57%). 1H NMR (400 MHz, DMSO-*) δ ppm 0.41 - 0.54 (m, 2 H), 0.73 - 0.89 (m, 2 H), 1.40 - 1.54 (m, 1 H), 1.63 - 1.78 (m, 3 H), 1.78 - 1.92 (m, 1 H), 1.91 - 2.04 (m, 2 H), 2.04 - 2.20 (m, 2 H), 2.87 - 3.04 (m, 7 H), 3.08 - 3.19 (m, 2 H), 3.43 (q, 7=6.41 Hz, 2 H), 6.89 (t, 7=5.95 Hz, 1 H), 7.30 (d, 7=8.70 Hz, 2 H), 7.63 (s, 1 H), 7.70 (t, 7=5.72 Hz, 1 H), 7.81 (d, 7=8.70 Hz, 2 H), 9.14 (s, 1 H) 437 [M+H]+.
Examples 181 - 183
The following examples were prepared analogously to Example 180 (the general structure is shown below followed by the tabulated examples).
Figure imgf000224_0001
Figure imgf000224_0002
a: HPLC column: 4.6x50mm (5μm) C- 18 Phenomenex Gemini-NX; flow rate: 2ml/min; Run time: 4.6 min: Solvent A: 0.1% Ammonium Hydroxide in water, Solvent B: Methanol; Gradient - 10- 100%B; Gradient time: 3.5min: b: HPLC column: 21.2xl00mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 40ml/min: gradient time: 7.5min, run time: 9 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: 0.1% ammonium hydroxide is added to the water
Example 184 Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[4-(2-oxo-pyrrolidin-l-yl)-phenylamino]- pyrimidin-4-ylamino}-propyl)-amide. xHCl
A solution of Intermediate 8 (50 mg, 0.16 mmol), l-(4-amino-phenyl)-pyrrolidin-2-one (80 mg, 0.49 mmol) and glacial AcOH (cat.) in n-butanol was heated at 150 0C in the microwave for 35 min. The resulting precipitate was filtered and recrystalised from 1 :1 MeOH:H2O to give the desired product (6.2 mg, 9%). 1H NMR (400 MHz, DMSO-J6) δ ppm 0.53 - 0.60 (m, 2 H), 0.86 - 0.94 (m, 2 H)1 1.49 - 1.59 (m, 1 H), 1.66 - 1.78 (m, 3 H), 1.78 - 1.91 (m, 1 H), 1.91 - 2.01 (m, 2 H), 2.02 - 2.14 (m, 4 H), 2.95 (quin, 7=8.24 Hz, 1 H), 3.05 - 3.15 (m, 2 H), 3.46 (q, 7=6.41 Hz, 2 H), 3.84 (t, /=7.10 Hz, 2 H), 7.50 - 7.61 (m, 3 H), 7.65 - 7.72 (m, 2 H), 7.77 (t, 7=5.72 Hz, 1 H), 8.59 (br. s, 1 H), 10.28 (s, 1 H), 11.91 (br. s, 1 H); m/z (ES+APCI)+ : 449 [M+H]+.
Example 185 Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(6-oxazol-5-yl-pyridin-3-ylamino)-pyrimidin-4- ylamino]-propyl}-amide
Figure imgf000225_0002
Prepared analogously to Example 172 from Intermediate 8 (70 mg, 0.23 mmol) and Intermediate 64 (35 mg, 0.23 mmol) to give the desired product as a white solid (9 mg, 9 %). 1H NMR (400 MHz, DMSCW6) δ ppm 0.41 - 0.56 (m, 2 H), 0.76 - 0.92 (m, 2 H), 1.48 (br. s, 1 H), 1.66 - 1.76 (m, 3 H), 1.83 (d, 7=10.07 Hz, 1 H), 1.97 (dd, J=8.70, 3.21 Hz, 2 H), 2.03 - 2.17 (m, 2 H), 2.97 (t, 7=8.24 Hz, 1 H), 3.09 - 3.20 (m, 2 H), 3.44 (d, 7=5.95 Hz, 2 H), 6.97 (s, 1 H), 7.58 (s, 1 H), 7.63 - 7.69 (m, 2 H), 7.71 (s, 1 H), 8.38 (dd, 7=8.93, 2.52 Hz, 1 H), 8.44 (s, 1 H), 8.93 (d, 7=2.29 Hz, 1 H), 9.38 (s, 1 H); m/z (ES+APCI)+ : 434 [M+H]+.
Examples 186-191
Examples 186-191 were prepared analogously to Example 172 (the general structure is shown below followed by the tabulated examples).
Figure imgf000226_0001
Figure imgf000226_0002
Figure imgf000227_0001
Figure imgf000228_0002
*HPLC column: 21.2xl00mm (lOμm) C-18 Phenomenex Gemini; flow rate: 20ml/min; run time: 10 min; gradient at start: 10% methanol and 90% water, gradient at finish: 100% methanol and 0% water; as buffer: ammonium bicarbonate (10 mmol) and ammonium hydroxide is added to the water.
Example 192
Cyclobutanecarboxylic acid (3-{5-cyclopropyl-2-[l-(tetrahydro-pyran-4-yl)-lH-pyrazol-4- ylamino]-pyrimidin-4-ylamino}-propyl) -amide
Figure imgf000228_0001
Prepared analogously to Example 91 using Intermediate 8 and l-(tetrahydro-pyran-4-yl)-lH- pyrazol-4-ylamine to provide the product as an off-white solid (18 mg, 32%). 1H NMR (400 MHz, DMSCW6) δ ppm 0.40 - 0.46 (m, 2 H), 0.77 - 0.84 (m, 2 H), 1.40 - 1.47 (m, 1 H), 1.65 - 1.78 (m, 3 H), 1.80 - 2.03 (m, 7 H), 2.06 - 2.17 (m, 2 H), 2.98 (quin, J=8.36 Hz, 1 H), 3.09 - 3.18 (m, 2 H), 3.37 - 3.49 (m, 4 H), 3.94 (dd, J=I 1.22, 3.43 Hz, 2 H), 4.27 - 4.36 (m, 1 H), 6.70 - 6.77 (m, 1 H), 7.45 (s, 1 H), 7.55 (s, 1 H), 7.71 (t, /=5.72 Hz, 1 H), 7.86 (s, 1 H), 8.69 (br. s, 1 H); m/z (ES+APCI)+: 440 [M+H]+.
Example 193
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2-(l -cyclopropylmethyl-1 H-indazol-6-ylamino)- pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000229_0001
Prepared analogously to Example 90 using Intermediate 8 and Intermediate 73 to provide the product as a white foamy solid (35 mg, 59%). 1H NMR (400 MHz, DMSO-^6) δ ppm 0.32 - 0.37 (m, 2 H), 0.43 - 0.52 (m, 4 H), 0.81 - 0.87 (m, 2 H), 1.19 - 1.29 (m, 1 H), 1.46 - 1.53 (m, 1 H), 1.66 - 1.87 (m, 4 H), 1.90 - 1.99 (m, 2 H), 2.03 - 2.14 (m, 2 H), 2.88 - 2.97 (m, 1 H), 3.15 (q, J=6.41 Hz, 2 H), 3.53 (q, J=6.26 Hz, 2 H), 4.15 (d, 7=6.87 Hz, 2 H), 6.89 (t, 7=5.72 Hz, 1 H), 7.19 - 7.23 (m, 1 H), 7.53 (d, 7=9.16 Hz, 1 H), 7.65 - 7.71 (m, 2 H), 7.84 (s, 1 H), 8.42 (s, 1 H), 9.16 (s, 1 H); m/z (ES+APCI)+: 460 [M+H]+.
Example 194
Cyclobutanecarboxylic acid {3-[5-cyclopropyl-2'(2-cyclopropylmethyl-2H-indazol-6-ylamino)- pyrimidin-4-ylamino]-propyl}-amide
Figure imgf000230_0001
Prepared analogously to Example 90 using Intermediate 8 and Intermediate 74 to provide the product as an off-white solid (28 mg, 47%). 1H NMR (400 MHz, DMSO-cfe) δ ppm 0.40 - 0.58 (m, 6 H), 0.80 - 0.86 (m, 2 H), 1.31 - 1.41 (m, 1 H), 1.44 - 1.51 (m, 1 H), 1.66 - 1.87 (m, 4 H), 1.88 - 1.99 (m, 2 H), 2.05 - 2.16 (m, 2 H), 2.90 - 2.99 (m, 1 H), 3.18 (q, /=6.41 Hz, 2 H), 3.43 - 3.51 (m, 2 H), 4.19 (d, 7=7.33 Hz, 2 H), 6.88 (t, 7=5.95 Hz, 1 H), 7.16 - 7.20 (m, 1 H), 7.49 (d, 7=9.16 Hz, 1 H), 7.64 (s, 1 H), 7.80 (t, 7=5.95 Hz, 1 H), 8.21 (s, 1 H), 8.33 (s, 1 H), 8.91 (s, 1 H); m/z (ES+APCI)+: 460 [M+H]+.
Examples 195-208
Examples 195-208 were prepared analogously to Example 91 (the general structure is shown below followed by the tabulated examples) using intermediate 8 and the appropriate amines. The amines for Examples 202, 203 and 205 were prepared analogously to Intermediate 70.
Figure imgf000230_0002
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
ΗPLC column: 21.2x100mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 40ml/min; Run time: 9.0 min: Solvent A: 0.1% Trifluoro acetic acid in water, Solvent B: Methanol; Gradient - 10-
100%B; Gradient time: 7.5min. bHPLC column: 4.6x50mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 2ml/min; Run time:
4.6 min: Solvent A: 0.1% Trifluoro acetic acid in water, Solvent B: Methanol; Gradient - 10-
100%B; Gradient time: 3.5min.
ΗPLC column: 4.6x50mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 2ml/min; Run time:
4.6 min: Solvent A: 0.1% Formic acid in water, Solvent B: Methanol; Gradient - 10-100%B;
Gradient time: 3.5min. dHPLC column: 4.6x50mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 2ml/min; Run time:
4.6 min: Solvent A: 0.1 % Ammonium Hydroxide in water, Solvent B: Methanol; Gradient - 10-
100%B; Gradient time: 3.5min.
ΗPLC column: 4.6x50mm (5μm) C-18 Xbridge; flow rate: 3ml/min; Run time: 3.2 min: Solvent
A: 0.1% Ammonium Hydroxide in water Solvent B: Acetonitrile; Gradient - 10-100%B; Gradient time: 2.35min.
Examples 209-214
Examples 209-214 were prepared analogously to Example 90 (the general structure is shown below followed by the tabulated examples) using Intermediate 8 and the appropriate amines.
Figure imgf000234_0001
Figure imgf000234_0002
ΗPLC column: 4.6x50mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 2ml/min; Run time: 4.6 min: Solvent A: 0.1% Trifluoro acetic acid in water, Solvent B: Methanol; Gradient - 10- 100%B; Gradient time: 3.5min. bHPLC column: 4.6x50mm (5μm) C-18 Phenomenex Gemini-NX; flow rate: 2ml/min; Run time: 4.6 min: Solvent A: 0.1% Formic acid in water, Solvent B: Methanol; Gradient - 10-100%B; Gradient time: 3.5min.
ΗPLC column: 4.6x50mm (5μm) C-18 Xbridge; flow rate: 3ml/min; Run time: 3.2 min: Solvent A: 0.1% Ammonium Hydroxide in water Solvent B: Acetonitrile; Gradient - 10-100%B; Gradient time: 2.35min.
Results
All compounds exemplified below in Table 1 have IC5O values against TBKl of 10 μM or better. The tables below shows a potency score for each compound (*** = TBKl IC50 <100 nM; * = TBKl IC50 between 100 nM and 1 μM; * = TBKl IC50 between 1 μM and 10 μM):
Table 1:
Figure imgf000235_0001
Figure imgf000236_0001
Figure imgf000236_0002
Figure imgf000237_0001
Table 2 below shows data for selected examples against a panel of different kinases, where the residual kinase activity, in %, at two concentrations of the compounds is expressed
Table 2:
Figure imgf000237_0002
Figure imgf000238_0001
Figure imgf000239_0001
Comparative data
Effect of 5-substituent on the pyrimidine ring on TBKl activity and selectivity over other protein kinases.
Summary
The following comparative data demonstrate that the 5-cyclopropyl pyrimidine derivatives exemplified in our patent show a unique and optimal balance of activity against the target enzyme versus selectivity over other protein kinases. The effect of substituting the 5-halo substituent on the pyrimidine ring with a cyclopropyl group was investigated and the 5-cyclopropyl derivatives showed improved kinase selectivity compared to their 5-halo counterparts. In addition to the 5- cyclopropyl derivatives, a range of linear and branched alkyl, as well as cyclopropylmethyl analogues were investigated (Table ) and were found to have significantly reduced activity against the target enzyme (TBKl) compared to Example 23. Without wishing to be bound by theory, it is believed that the unique combination of steric and electronic properties of the cyclopropyl group account for these surprising and unpredictable results.
Comparison of a 5-halo pyrimidine with a cyclopropyl derivative
Table compares the activity of Example 23 with cyclobutanecarboxylic acid {3-[5-bromo-2-(3- fluoro-phenylamino)-pyrimidin-4-ylamino]-propyl}-arnide (Compound A)13, against a panel of different kinases, where the residual kinase activity, in %, at two concentrations of the compounds is expressed. The higher the percentage residual activity, the lower the degree of inhibition of the kinase. The Gini coefficient (value between 0 and 1) has recently been used to quantify the selectivity profile of compounds against large numbers of kinases, with higher values indicating a higher degree of selectivity.12 The data suggest that whilst both compounds show a similar inhibition of TBKl, replacement of the 5-bromo substituent on the pyrimidine ring with a cyclopropyl group (Example 23) results in a significant improvement of selectivity across this kinase panel. This is also reflected in the Gini coefficients, with Compound A giving a significantly lower value than Example 23.
Table 3: Selectivity data against a panel of different kinases
Figure imgf000240_0001
Figure imgf000241_0001
Table 4: Structures of Example 23 and compound A
Figure imgf000242_0001
Comparison of a 5-cycIopropylpyrimidiπe with isopropyl, cyclopropylmethyl and ethyl derivatives
Table 5 compares the IC5O values against TBKl of Example 23 with cyclobutanecarboxylic acid {3-[2-(3-fluoro-phenylamino)-5-isopropyl-pyrimidin-4-ylamino]-propyl}-amide (Compound B), cyclobutanecarboxylic acid {3-[5-cyclopropylmethyl-2-(3-fluoro-phenylamino)-pyrimidin-4- ylamino]-propyl}-amide (Compound C) and cyclobutanecarboxylic acid {3-[5-ethyl-2-(3-fluoro- phenylamino)-pyrimidin-4-ylamino]-propyl} -amide (Compound D). These data indicate that the 5- cyclopropyl derivative (Example 23) was found to be >250-fold more active than both the isopropyl derivative (Compound B) or cyclopropylmethyl derivative (Compound C). The ethyl derivative (Example D) shows a 10-fold loss in activity compared to Example 23.
Table 5: IC50 values against TBKl of Example 23, Compound B13 and Compound C13
Figure imgf000243_0001
REFERENCES & NOTES
1. Rezaie, T., Child, A., Hitchings, R., Brice, G., Miller, L., Coca-Prados, M., Heon, E., Krupin, T., Ritch, R., Kreutzer, D., Crick, R.P. and Sarfarazi, M. (2002) Adult-onset primary open- angle glaucoma caused by mutations in optineurin. Science 295, 1077-1079.
2. Sarfarazi, M. and Rezaie, T. (2003) Optineurin in primary open angle glaucoma. Ophthalmol Clin North Am 16, 529-541.
3. Tezel, G. and Wax, M.B. (2000) Increased production of tumour necrosis factor-alpha by glial cells exposed to stimulated ischemia or elevated hydrostatic pressure induces apoptosis in cocultured retinal ganglion cells. J Neurosci 20, 8693-8700.
4. Yuan, L. and Neufeld, A.H. (2000) Tumor necrosis factor-alpha: a potentially neurodestructive cytokine produced by glia in the human glaucomatous optic nerve head. GHa 32, 42-50.
5. Perry et al (J Exp Med 199, 1651-1658, 2004) compared the role of TBKl in interferon responses induced by a number of stimuli. TBK-/- mice were deficient in their ability to up regulate IFN beta production.
6. McWhirter et al (PNAS 101, 233 238, 2004) Demonstrate that induction of type I interferon and related genes depends on TBKl. They also show that IKKepsilon and TBKl directly phosphorylate serine residues that are critical for IRF3 activation.
7. Hemmi et al (J Exp Med 199, 1641-1650, 2004) indicate that TBKl and IKK are essential for the activation of IFN beta and IFN inducible genes.
8. Davies, S. P., Reddy, H., Caivano, M. and Cohen, P. (2000) Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J 351, 95-105.
9. Bain, J., McLauchlan, H., Elliott, M. and Cohen, P. (2003) The specificities of protein kinase inhibitors: an update. Biochem J 371, 199-204.
10. Schwamborn, K., Weil, R., Courtois, G., Whiteside, S.T. and Israel, A. (2000) Phorbol esters and cytokines regulate the expression of the NEMO-related protein, a molecule involved in a NF-kappa B-independent pathway. J Biol Chem 275, 22780-22789.
11. Morton, S., Hesson, L., Peggie, M. and Cohen, P. (2008) Enhanced binding of TBKl by an optineurin mutant that causes a familial form of primary open angle glaucoma. FEBS Letters 582, 997-1002.
12. P.P. Graczyk, J. Med. Chem. 50 (2007), p. 5773 13. Compounds A, B and D are generically claimed in WO 2004/048343 and can be prepared analogously to procedures outlined in this patent application; compound C is generically claimed in WO 2002/064586. Spectrocopic data for compound C: 1H NMR (400 MHz, DMSO- d&) δ ppm 0.12 - 0.25 (m, 2 H), 0.35 - 0.59 (m, 2 H), 0.94 - 1.12 (m, 1 H), 1.69 - 1.81 (m, 3 H), 1.85 - 1.95 (m, 1 H), 1.96 - 2.06 (m, 2 H), 2.08 - 2.19 (m, 2 H), 2.29 (d, 7=6.9 Hz, 2 H), 2.99 (quin, 7=8.2 Hz, 1 H), 3.15 (q, 7=6.6 Hz, 2 H), 3.40 - 3.55 (m, 2 H), 6.66 (td, 7=8.5, 2.3 Hz, 1 H), 6.82 (t, 7=5.7 Hz, 1 H), 7.22 - 7.32 (m, I H), 7.48 (d, 7=9.6 Hz, 1 H), 7.70 (t, 7=5.7 Hz, 1 H), 7.81 (s, 1 H), 7.91 (dt, 7=13.3, 2.3 Hz, 1 H), 9.17 (s, 1 H); m/z (ES+APCI)+: 398 [M+H]+

Claims

1. A compound of formula (I), or a pharmaceutically acceptable salt or ester thereof,
Figure imgf000246_0001
(I) wherein: R1 is Cs-g-cycloalkyl;
X is O, NR7 or C3^-heterocycloalkyl;
R2 is aryl, heteroaryl, fused or unfused aryl-C3-6-heterocycloalkyl or fused or unfused heteroaryI-C3. 6-heterocycloalkyl, each of which is optionally substituted by one or more substitutents selected from aryl, heteroaryl, Ci-β-alkyl, C3.7-cycloalkyl and a group A, wherein said Ci-6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl, R10 and a group A, said heteroaryl group is optionally substituted by one or more R10 groups; and wherein said C3-5- heterocycloalkyl group optionally contains one or more groups selected from oxygen, sulfur, nitrogen and CO;
R3 is Ci-6-alkyl optionally substituted by one or more substituents selected from aryl, heteroaryl, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7SO2R6, -NR7COOR7, -CONR4R5, C3-5-heterocycloalkyl and
Figure imgf000246_0002
wherein said aryl, heteroaryl and C^-heterocycloalkyl groups are each optionally substituted by one or more substituents selected from
Figure imgf000247_0001
and a group A, wherein said -C)-6-alkyl group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A;
A is selected from halogen, hydroxyl, cyano, trifluoromethyl, alkoxy, -NO2, -NH2, -NR4R5, -OR6, -NR7(CO)R6, -NR7(CO)NR4R5, -NR7COOR7, -NR7(SO2)R6, - CO2H, -NR7CSO2)NR4R5, -COOR7, -CONR4R5, COR6, SO2NR4R5 and -SO2CH3;
each R4 and R5 is independently selected from hydrogen, C3-7-cycloalkyl, aryl, heteroaryl, Ci-6-alkyl and a C^-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more R10 groups, wherein said Ci_5-alkyl is optionally substituted by one or more substituents selected from halogen, cyano, hydroxyl, aryl, heteroaryl, -NR8R9, -NR7(CO)R6, NR7COOR6, -NR7(SO2)R6, -COOR6, -CONR8R9, OR10, -SO2R6 and a C3-6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO and optionally substituted by one or more or R10 groups; or
R4 and R5 together with the N to which they are attached form a C3-6-heterocycloalkyl ring optionally further containing one or more groups selected from oxygen, sulfur, nitrogen and CO, wherein said C3-6-heterocycloalkyl ring may be saturated or unsaturated and is optionally substituted with one or more groups selected from NR8R9 and R10 groups;
each R6 is independently selected from Ci-6-alkyl, C3-7 cycloalkyl, C4.7-heterocycloalkyl, aryl and heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9;
each R7 is selected from hydrogen,
Figure imgf000247_0002
and C3.7-cycloalkyl, wherein said
Figure imgf000247_0003
is optionally substituted by one or more halogens;
each of R8 and R9 is independently selected from hydrogen and
Figure imgf000247_0004
wherein said
Figure imgf000247_0005
group is optionally substituted by one or more halogens; or R8 and R9 together with the N to which they are attached form a C^-heterocycloalkyl ring optionally further containing one or more heteroatoms selected from oxygen and sulfur, wherein said C4-6-heterocycloalkyl ring is optionally substituted by one or more R10 groups; and
each R10 is selected from halogen, C3.7-cycloalkyl and C^-alkyl optionally substituted by one or more halogens, wherein where R10 is
Figure imgf000248_0001
and two or more R10 groups are attached to the same carbon atom, the R10 groups may be linked to form a spiroalkyl group.
2. A compound according to claim 1 wherein R1 is cyclopropyl or cyclobutyl.
3. A compound according to claim 1 or claim 2 wherein R1 is cyclopropyl.
4. A compound according to any preceding claim wherein X is NH.
5. A compound according to any preceding claim wherein R2 is an optionally substituted aryl or heteroaryl group selected from the following:
Figure imgf000249_0001
Figure imgf000249_0002
6. A compound according to any preceding claim wherein R2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from C^-alkyl and a group A, wherein said
Figure imgf000249_0003
group is optionally substituted by one or more substituents selected from aryl, heteroaryl and a group A, and wherein A is selected from halogen, OH, CN, CF3, -NH2, -NR4R5, -OR6, NR7(CO)R6, -NR7COOR7, -NR7(SO2)R6, -COOH, -COOR7 and CONR4R5.
7. A compound according to any preceding claim wherein R2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from
Figure imgf000249_0004
halogen, CN, NHCO-C 1-6-alkyl, CF3, COOH, CONH2, OH, NH2, NHSO2-C1-6-alkyl, 0-CF3, - NHCOO-C i-5-alkyl, -CO2-C]^-alkyl, -N(Ci.6-alkyl)2, 4-methylpiperazin-l-yl, (4-methylpiperazin- l-yl)-CO-, (N-morpholinyl)-(CH2)p(O)q-, (imidazol-l-yl)-(CH2)p- where q is O, 1, 2 or 3 and each p is independently 1, 2 or 3 and NR4R5, wherein R4 and R5 and the nitrogen to which they are attached form a C3-6-heterocycloalkyl ring optionally containing a CO group.
8. A compound according to any preceding claim wherein R2 is an aryl or heteroaryl group each of which is optionally substituted by one or more substituents selected from Me, Cl, F, CN, NHCOMe, CF3, COOH, CONH2, OH, NH2, NHSO2Me, OCF3, -NHCOO1Bu, -CO2Me, -NMe2, 4- methylpiperazin-1-yl, N-morpholinyl, (4-methylpiperazin-l-yl)-C0-, (N-morpholinyl)-CH2CH2O-, (imidazol-l-yl)-CH2- and
Figure imgf000250_0001
9. A compound according to claim 1 wherein R2 is:
(a) a phenyl group substituted by one or more A groups, wherein A is preferably selected from CF3, halogen, CN, NHSO2Me, SO2NR4R5, NR4R5, OR6, COOR7, NR7COOR7, NR7COR6, CONR4R5, NR7CONR4R5 and NR7SO2R6;
(b) a phenyl group substituted by one or more
Figure imgf000250_0002
groups, each of which in turn is optionally substituted by one or more groups selected from heteroaryl and A, wherein the heteroaryl group is preferably selected from imidazolyl and triazolyl, and the A group is preferably selected from CONR4R5, NR4R5, OR6, COOR7 and CN;
(c) a phenyl group substituted by one or more heteroaryl groups, wherein the heteroaryl group is preferably selected from pyrimidinyl, tetrazolyl, pyridinyl, pyrazolyl, oxazolyl and triazolyl;
(d) a pyridyl group substituted by one or more A groups, wherein the A group is preferably selected from NR4R5, halo and OR6;
(e) a pyridyl group substituted by a heteroaryl group, wherein the heteroaryl group is preferably selected from pyrazolyl, pyrimidinyl and pyridinyl;
(f) a pyridyl group substituted by a Ci_5-alkyl group, wherein said Ci.6-alkyl group is in turn optionally substituted with one or more substituents selected from NR4R5 and OR6; (g) an optionally substituted fused aryl-C3^-heterocycloalkyl or fused heteroaryl-C^- heterocycloalkyl, preferably selected from the following:
Figure imgf000251_0001
10. A compound according to any preceding claim wherein R3 is C^-alkyl optionally substituted by one or more substituents selected from heteroaryl, -NR4R5, -NR7(CO)R6, -NR7COOR7, C^-heterocycloalkyl and
Figure imgf000251_0002
1 1. A compound according to any preceding claim wherein R3 is C^-alkyl substituted by -NR7(CO)R6.
12. A compound according to claim 11 wherein R7 is H and R6 is selected from CWalkyl, C3.7 cycloalkyl, C4.7-heterocycloalkyl, heteroaryl, each of which may be optionally substituted by one or more substituents selected from halogen, R10 and -NR8R9.
13. A compound according to claim 12 wherein R6 is selected from thienyl, cyclopentyl, CH2- cyclopentyl, isopropyl, pyrazolyl, cyclohexyl, thiazolyl, oxazolyl, furanyl, CF3, imidazolyl, cyclopropyl, CH2-cyclopropyl, cyclobutyl, triazolyl, pyrrolyl, tetrahydrofuranyl, CH2NMe2 and iso- oxazolyl.
14. A compound according to any one of claims 1 to 10 wherein R3 is C^-alkyl substituted by a heteroaryl selected from pyrazolyl, tetrazolyl and triazolyl.
15. A compound according to any one of claims 1 to 10 wherein R3 is CM-alkyl substituted by a -NR4R5 group, wherein R4 and R5 together with the N to which they are attached form a C3-6- heterocycloalkyl ring optionally further containing one or more CO groups, wherein said C3-6- heterocycloalkyl ring is optionally substituted with one or more R10 groups.
16. A compound according to any one of claims 1 to 10 wherein R3 is
Figure imgf000252_0001
substituted by one of the following groups:
Figure imgf000252_0002
each of which may be optionally substituted with one or more R10 groups.
17. A compound according to any one of claims 1 to 10 wherein R3 is d^-alkyl substituted by a C3-6-heterocycloalkyl group, wherein said C^-heterocycloalkyl group is optionally substituted by one or more A groups.
18. A compound according to any preceding claim wherein R3 is an optionally substituted C3- alkyl group.
19. A compound according to claim 1 which is selected from the following:
Figure imgf000253_0001
Figure imgf000254_0001
Figure imgf000255_0001
Figure imgf000256_0001
Figure imgf000257_0001
Figure imgf000258_0001
105 106 107 108 109 110 111
Figure imgf000259_0001
Figure imgf000260_0001
Figure imgf000261_0001
Structure Example Structure Example
Example 145 Example 153
Figure imgf000262_0001
KX. JC rcέ; Example 146 CX. Example 154
C Example 147 O Example 155
C Example 148 Example 156
Example 149 Example 157
Example 150 OL Example 158
Figure imgf000262_0002
QX f Example 151 >o Example 159
Ϋ Example 152 Example 160
169 170 171 172 173 174 175 176
Figure imgf000263_0001
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
20. A pharmaceutical composition comprising at least one compound according to any one of claims 1 to 19 and a pharmaceutically acceptable carrier, diluent or excipient.
21. A compound according to any one of claims 1 to 19 for use in medicine.
22. A compound according to any one of claims 1 to 19 for use in treating a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
23. Use of a compound according to any one of claims 1 to 19 in the preparation of a medicament for treating or preventing a disorder selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, psoriasis, artherosclerosis, retinopathy, osteoarthritis, endometriosis and chronic inflammation.
24. Use according to claim 23 wherein the compound is administered in an amount sufficient to inhibit a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR and IKKepsilon.
25. Use of a compound according to any one of claims 1 to 19 in the preparation of a medicament for the prevention or treatment of a disorder caused by, associated with or accompanied by any abnormal kinase activity, wherein the kinase is selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, IKKepsilon and combinations thereof.
26. Use according to claim 25 wherein the kinase is selected from TBKl, PDKl, ERK8, MARK3, and IKKepsilon and combinations thereof.
27. Use according to claim 25 or claim 26 wherein the kinase is selected from TBKl and PDKl.
28. Use according to any one of claims 25 to 27 wherein the kinase is TBKl.
29. Use according to any one of claims 25 to 28 wherein the disorder is selected from cancer, septic shock, neurodegenerative diseases, Alzheimer's disease, diseases of the eye, including Primary open Angle Glaucoma (POAG), hyperplasia, rheumatoid arthritis, autoimmune diseases, artherosclerosis, retinopathy, osteoarthritis, fibrotic diseases, endometriosis and chronic inflammation.
30. Use according to claim 23 or claim 29 wherein the cancer is selected from papilloma, blastoglioma, Kaposi's sarcoma, melanoma, lung cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, astrocytoma, head cancer, neck cancer, skin cancer, liver cancer, bladder cancer, breast cancer, lung cancer, uterus cancer, prostate cancer, testis carcinoma, colorectal cancer, thyroid cancer, pancreatic cancer, gastric cancer, hepatocellular carcinoma, leukemia, lymphoma, Hodgkin's disease and Burkitt's disease; the disease of the eye is selected from glaucoma, primary open angle glaucoma (POAG), normal tension glaucoma (NTG) and low tension laucoma (LTG); the auto-immune disease is selected from psoriasis, alopecia and multiple sclerosis; and the fibrotic disease is cirrhosis of the liver.
31. A method of treating a mammal having a disease state alleviated by the inhibition of a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, wherein the method comprises administering to a mammal a therapeutically effective amount of a compound according to any one of claims 1 to 19.
32. A method according to claim 31 wherein the disease state is alleviated by the inhibition of a kinase selected from TBKl, PDKl, ERK8, MARK3, and IKKepsilon.
33. A method according to claim 31 or claim 32 wherein the disease state is alleviated by the inhibition of TBKl or PDKl.
34. A method according to any one of claims 31 to 33 wherein the disease state is alleviated by the inhibition of TBKl.
35. A method according to any one of claims 31 to 34 wherein the mammal is a human.
36. Use of a compound according to any one of claims 1 to 19 in an assay for identifying further candidate compounds capable of inhibiting one or more kinases selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon.
37. Use according to claim 36 wherein said assay is a competitive binding assay.
38. Use according to claim 37 wherein said competitive binding assay comprises contacting a compound according to any one of claims 1 to 19 with a kinase selected from TBKl, MKKl, ERK8, RSKl, RSK2, PDKl, S6K1, MNK2, PHK, CHKl, CHK2, GSK3beta, CDK2, MARK3, MELK, IRR, VEG-FR, and IKKepsilon, and a candidate compound and detecting any change in the interaction between the compound according to any one of claims 1 to 19 and the kinase.
39. A process for preparing a compound of formula IV, wherein R1, R2, R3 and R7 are as defined in any one of claims 1 to 19, said process comprising the steps of:
Figure imgf000269_0001
(X=O1 S)
Ia II πi rv
(i) converting a compound of formula Ia to a compound of formula II, where each LG is independently a leaving group; (ii) reacting said compound of formula II with an amine of formula IV to form a compound of formula III; (iii) reacting said compound of formula III with an amine of formula VII to form a compound of formula IV.
40. A process for preparing a compound o ff ffoorrrmr ula XIX, wherein R1, R2 and R3 are as defined in any one of claims 1 to 19, said process comprising the steps of:
Figure imgf000270_0001
(X=O, S)
Ia II XVIII XIX
(i) converting a compound of formula Ia to a compound of formula II, where each LG is independently a leaving group; (ii) reacting said compound of formula II with an amine of formula XVII to form a compound of formula XVIII; (iii) reacting said compound of formula XVIII with an amine of formula VII to form a compound of formula XIX.
41. A combination comprising a compound according to any one of claims 1 to 19 and a further therapeutic agent.
42. A pharmaceutical composition according to claim 20 which further comprises a second therapeutic agent.
PCT/GB2009/000878 2008-04-02 2009-04-02 Pyrimidine derivatives capable of inhibiting one or more kinases WO2009122180A1 (en)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US7276408P 2008-04-02 2008-04-02
GB0806005A GB0806005D0 (en) 2008-04-02 2008-04-02 Compound
GB0806005.5 2008-04-02
US61/072,764 2008-04-02
GB0812580.9 2008-07-09
GB0812580A GB0812580D0 (en) 2008-07-09 2008-07-09 Compound
GB0822635A GB0822635D0 (en) 2008-12-11 2008-12-11 Compound
GB0822635.9 2008-12-11

Publications (1)

Publication Number Publication Date
WO2009122180A1 true WO2009122180A1 (en) 2009-10-08

Family

ID=40790560

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2009/000878 WO2009122180A1 (en) 2008-04-02 2009-04-02 Pyrimidine derivatives capable of inhibiting one or more kinases

Country Status (2)

Country Link
US (1) US20100056524A1 (en)
WO (1) WO2009122180A1 (en)

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012161877A1 (en) 2011-05-23 2012-11-29 Merck Patent Gmbh Pyridine-and pyrazine derivatives
US8354420B2 (en) 2010-06-04 2013-01-15 Genentech, Inc. Aminopyrimidine derivatives as LRRK2 inhibitors
WO2013024282A2 (en) 2011-08-15 2013-02-21 Domainex Limited Compounds and their uses
WO2013034238A1 (en) 2011-09-09 2013-03-14 Merck Patent Gmbh Benzonitrile derivatives as kinase inhibitors
JP2013514370A (en) * 2009-12-17 2013-04-25 メルク・シャープ・エンド・ドーム・コーポレイション Aminopyrimidines as Syk inhibitors
DE102011119127A1 (en) 2011-11-22 2013-05-23 Merck Patent Gmbh 3-Cyanaryl-1H-pyrrolo [2,3-b] pyridine derivatives
EP2597955A1 (en) * 2010-07-30 2013-06-05 OncoTherapy Science, Inc. Quinoline derivatives and melk inhibitors containing the same
WO2013117285A1 (en) 2012-02-09 2013-08-15 Merck Patent Gmbh Furo [3, 2 - b] - and thieno [3, 2 - b] pyridine derivatives as tbk1 and ikk inhibitors
WO2013136070A1 (en) 2012-03-13 2013-09-19 University Court Of The University Of Dundee A sik inhibitor for use in a method of treating an inflammatory and/or immune disorder
US8735417B2 (en) 2009-12-17 2014-05-27 Merck Sharp & Dohme Corp. Aminopyrimidines as Syk inhibitors
US8815882B2 (en) 2010-11-10 2014-08-26 Genentech, Inc. Pyrazole aminopyrimidine derivatives as LRRK2 modulators
JP2014533737A (en) * 2011-11-29 2014-12-15 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 2- (Phenyl or pyrid-3-yl) aminopyrimidine derivatives as kinase LRRK2 modulators for the treatment of Parkinson's disease
CN105153131A (en) * 2015-09-21 2015-12-16 马彦丽 Medicine combination for treating human neuroglioma during nursing
US9249114B2 (en) 2011-05-23 2016-02-02 Merck Patent Gmbh Thiazole derivatives
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
JP2016185953A (en) * 2011-06-10 2016-10-27 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2018030881A3 (en) * 2017-01-03 2018-03-29 윤성문 Use of thyroid hormone for treatment of choroidal disease
WO2018114999A1 (en) * 2016-12-20 2018-06-28 Astrazeneca Ab Amino-triazolopyridine compounds and their use in treating cancer
US10040781B2 (en) 2014-09-26 2018-08-07 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10072001B2 (en) 2014-06-03 2018-09-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10265321B2 (en) 2014-08-08 2019-04-23 Dana-Farber Cancer Institute, Inc. Uses of salt-inducible kinase (SIK) inhibitors
US10287268B2 (en) 2014-07-21 2019-05-14 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10316049B2 (en) 2015-12-17 2019-06-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10457691B2 (en) 2014-07-21 2019-10-29 Dana-Farber Cancer Institute, Inc. Macrocyclic kinase inhibitors and uses thereof
JP2020508341A (en) * 2017-02-23 2020-03-19 ドメイネクス リミテッド 5- (Pyrimidin-4-yl) -2- (pyrrolidin-1-yl) nicotinonitrile Compounds as IKKE, TBK1 and / or SIK2 Kinase Inhibitors
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10954242B2 (en) 2016-07-05 2021-03-23 The Broad Institute, Inc. Bicyclic urea kinase inhibitors and uses thereof
CN113493471A (en) * 2020-04-03 2021-10-12 山东轩竹医药科技有限公司 Heteroaromatic kinase inhibitors
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11241435B2 (en) 2016-09-16 2022-02-08 The General Hospital Corporation Uses of salt-inducible kinase (SIK) inhibitors for treating osteoporosis
CN114127057A (en) * 2019-05-10 2022-03-01 德西费拉制药有限责任公司 Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
US11285158B2 (en) 2017-02-28 2022-03-29 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
WO2022199561A1 (en) * 2021-03-23 2022-09-29 杭州阿诺生物医药科技有限公司 Hpk1 kinase inhibitor compound
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
WO2022251584A1 (en) * 2021-05-28 2022-12-01 St. Jude Children's Research Hospital, Inc. Methods and compositions for transduction of nk cells
US11518758B2 (en) 2019-05-10 2022-12-06 Deciphera Pharmaceuticals, Llc Heteroarylaminopyrimidine amide autophagy inhibitors and methods of use thereof
CN115650956A (en) * 2022-10-11 2023-01-31 深圳市龙华区中心医院 Small molecule compound for targeted inhibition of activity of human and mouse RIPK3 protein kinases
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11590134B2 (en) 2019-06-17 2023-02-28 Deciphera Pharmaceuticals, Llc Aminopyrimidine amide autophagy inhibitors and methods of use thereof
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2937345B1 (en) 2009-12-29 2018-03-21 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors
WO2012142329A1 (en) * 2011-04-12 2012-10-18 Myrexis, Inc. Compositions and therapeutic uses of ikk-related kinase epsilon and tankbinding kinase 1 inhibitors
JP6106685B2 (en) 2011-11-17 2017-04-05 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド Inhibitors of C-JUN-N-terminal kinase (JNK)
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
USRE48175E1 (en) 2012-10-19 2020-08-25 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
GB201311891D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compound
GB201311888D0 (en) 2013-07-03 2013-08-14 Glaxosmithkline Ip Dev Ltd Novel compounds
CA2927920A1 (en) 2013-10-18 2015-04-23 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (cdk7)
WO2015058126A1 (en) 2013-10-18 2015-04-23 Syros Pharmaceuticals, Inc. Heteroaromatic compounds useful for the treatment of prolferative diseases
WO2015164614A1 (en) 2014-04-23 2015-10-29 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
AU2015371251B2 (en) 2014-12-23 2020-06-11 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
CN107427521B (en) 2015-03-27 2021-08-03 达纳-法伯癌症研究所股份有限公司 Inhibitors of cyclin dependent kinases
WO2016197114A1 (en) 2015-06-05 2016-12-08 Arvinas, Inc. Tank-binding kinase-1 protacs and associated methods of use
CA2986441A1 (en) 2015-06-12 2016-12-15 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
EP4019515A1 (en) 2015-09-09 2022-06-29 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002096888A1 (en) * 2001-05-29 2002-12-05 Schering Aktiengesellschaft Cdk inhibiting pyrimidines, production thereof and their use as medicaments
WO2004048343A1 (en) * 2002-11-28 2004-06-10 Schering Aktiengesellschaft Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents
WO2008003766A2 (en) * 2006-07-06 2008-01-10 Boehringer Ingelheim International Gmbh 4-heter0cycl0alkylpyri(mi)dines, process for the preparation thereof and their use as medicaments
WO2008051547A1 (en) * 2006-10-23 2008-05-02 Cephalon, Inc. Fused bicyclic derivatives of 2,4-diaminopyrimidine as alk and c-met inhibitors
WO2008129380A1 (en) * 2007-04-18 2008-10-30 Pfizer Products Inc. Sulfonyl amide derivatives for the treatment of abnormal cell growth

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2332924A1 (en) * 2001-10-17 2011-06-15 Boehringer Ingelheim Pharma GmbH & Co. KG Pyrimidine derivates, medicaments comprising them, their use and process of their preparation
US20070203161A1 (en) * 2006-02-24 2007-08-30 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway
US20070032514A1 (en) * 2005-07-01 2007-02-08 Zahn Stephan K 2,4-diamino-pyrimidines as aurora inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002096888A1 (en) * 2001-05-29 2002-12-05 Schering Aktiengesellschaft Cdk inhibiting pyrimidines, production thereof and their use as medicaments
WO2004048343A1 (en) * 2002-11-28 2004-06-10 Schering Aktiengesellschaft Chk-, pdk- and akt-inhibitory pyrimidines, their production and use as pharmaceutical agents
WO2008003766A2 (en) * 2006-07-06 2008-01-10 Boehringer Ingelheim International Gmbh 4-heter0cycl0alkylpyri(mi)dines, process for the preparation thereof and their use as medicaments
WO2008051547A1 (en) * 2006-10-23 2008-05-02 Cephalon, Inc. Fused bicyclic derivatives of 2,4-diaminopyrimidine as alk and c-met inhibitors
WO2008129380A1 (en) * 2007-04-18 2008-10-30 Pfizer Products Inc. Sulfonyl amide derivatives for the treatment of abnormal cell growth

Cited By (100)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8551984B2 (en) 2009-12-17 2013-10-08 Merck Sharp & Dohme Corp. Aminopyrimidines as SYK inhibitors
US8735417B2 (en) 2009-12-17 2014-05-27 Merck Sharp & Dohme Corp. Aminopyrimidines as Syk inhibitors
JP2013514370A (en) * 2009-12-17 2013-04-25 メルク・シャープ・エンド・ドーム・コーポレイション Aminopyrimidines as Syk inhibitors
JP2013155182A (en) * 2009-12-17 2013-08-15 Merck Sharp & Dohme Corp AMINOPYRIMIDINES AS Syk INHIBITOR
US8354420B2 (en) 2010-06-04 2013-01-15 Genentech, Inc. Aminopyrimidine derivatives as LRRK2 inhibitors
EP2597955A1 (en) * 2010-07-30 2013-06-05 OncoTherapy Science, Inc. Quinoline derivatives and melk inhibitors containing the same
KR101826382B1 (en) 2010-07-30 2018-02-06 온코세라피 사이언스 가부시키가이샤 Quinoline derivatives and melk inhibitors containing the same
EP2597955A4 (en) * 2010-07-30 2014-02-19 Oncotherapy Science Inc Quinoline derivatives and melk inhibitors containing the same
US9120749B2 (en) 2010-07-30 2015-09-01 Oncotherapy Science, Inc. Quinoline derivatives and MELK inhibitors containing the same
JP2017008074A (en) * 2010-11-10 2017-01-12 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Pyrazole aminopyrimidine derivatives as lrrk2 modulators
US8815882B2 (en) 2010-11-10 2014-08-26 Genentech, Inc. Pyrazole aminopyrimidine derivatives as LRRK2 modulators
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9249114B2 (en) 2011-05-23 2016-02-02 Merck Patent Gmbh Thiazole derivatives
WO2012161877A1 (en) 2011-05-23 2012-11-29 Merck Patent Gmbh Pyridine-and pyrazine derivatives
JP2014518884A (en) * 2011-05-23 2014-08-07 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Pyridine- and pyrazine derivatives
EA023364B1 (en) * 2011-05-23 2016-05-31 Мерк Патент Гмбх Pyridine and pyrazine derivatives
US9273029B2 (en) 2011-05-23 2016-03-01 Merck Patent Gmbh Pyridine-and pyrazine derivatives
JP2016185953A (en) * 2011-06-10 2016-10-27 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Compositions and methods for the production of pyrimidine and pyridine compounds with btk inhibitory activity
JP2018115165A (en) * 2011-06-10 2018-07-26 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Compositions and methods for production of pyrimidine and pyridine compounds with btk inhibitory activity
WO2013024282A3 (en) * 2011-08-15 2013-05-16 Domainex Limited Inhibitor or down- regulator of the expression of one or both of tbk1 ikk-epsilon for use in the treatment of pi3kinase dependent cancer
WO2013024282A2 (en) 2011-08-15 2013-02-21 Domainex Limited Compounds and their uses
DE102011112978A1 (en) 2011-09-09 2013-03-14 Merck Patent Gmbh benzonitrile derivatives
WO2013034238A1 (en) 2011-09-09 2013-03-14 Merck Patent Gmbh Benzonitrile derivatives as kinase inhibitors
DE102011119127A1 (en) 2011-11-22 2013-05-23 Merck Patent Gmbh 3-Cyanaryl-1H-pyrrolo [2,3-b] pyridine derivatives
WO2013075785A1 (en) 2011-11-22 2013-05-30 Merck Patent Gmbh 3-cyanaryl-1h-pyrazolo[2.3-b]pyridine derivatives
JP2014533737A (en) * 2011-11-29 2014-12-15 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 2- (Phenyl or pyrid-3-yl) aminopyrimidine derivatives as kinase LRRK2 modulators for the treatment of Parkinson's disease
WO2013117285A1 (en) 2012-02-09 2013-08-15 Merck Patent Gmbh Furo [3, 2 - b] - and thieno [3, 2 - b] pyridine derivatives as tbk1 and ikk inhibitors
WO2013136070A1 (en) 2012-03-13 2013-09-19 University Court Of The University Of Dundee A sik inhibitor for use in a method of treating an inflammatory and/or immune disorder
US10233157B2 (en) 2012-03-13 2019-03-19 Dana-Farber Cancer Institute, Inc. SIK inhibitor for use in a method of treating an inflammatory and/or immune disorder
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10259811B2 (en) 2014-06-03 2019-04-16 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10072001B2 (en) 2014-06-03 2018-09-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10975058B2 (en) 2014-07-21 2021-04-13 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10287268B2 (en) 2014-07-21 2019-05-14 Dana-Farber Cancer Institute, Inc. Imidazolyl kinase inhibitors and uses thereof
US10457691B2 (en) 2014-07-21 2019-10-29 Dana-Farber Cancer Institute, Inc. Macrocyclic kinase inhibitors and uses thereof
US10265321B2 (en) 2014-08-08 2019-04-23 Dana-Farber Cancer Institute, Inc. Uses of salt-inducible kinase (SIK) inhibitors
US10040781B2 (en) 2014-09-26 2018-08-07 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10253019B2 (en) 2014-09-26 2019-04-09 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
CN105153131A (en) * 2015-09-21 2015-12-16 马彦丽 Medicine combination for treating human neuroglioma during nursing
US10316049B2 (en) 2015-12-17 2019-06-11 Gilead Sciences, Inc. Tank-binding kinase inhibitor compounds
US10954242B2 (en) 2016-07-05 2021-03-23 The Broad Institute, Inc. Bicyclic urea kinase inhibitors and uses thereof
US11725011B2 (en) 2016-07-05 2023-08-15 The General Hospital Corporation Bicyclic urea kinase inhibitors and uses thereof
US11241435B2 (en) 2016-09-16 2022-02-08 The General Hospital Corporation Uses of salt-inducible kinase (SIK) inhibitors for treating osteoporosis
US10407446B2 (en) 2016-12-20 2019-09-10 Astrazeneca Ab Amino-triazolopyridine compounds and their use in treating cancer
US11136340B2 (en) 2016-12-20 2021-10-05 Astrazeneca Ab Amino-triazolopyridine compounds and their use in treating cancer
CN110177791A (en) * 2016-12-20 2019-08-27 阿斯利康(瑞典)有限公司 Amino-triazolopyridine compound and its purposes in treating cancer
US11746118B2 (en) 2016-12-20 2023-09-05 Astrazeneca Ab Amino-triazolopyridine compounds and their use in treating cancer
EA037745B1 (en) * 2016-12-20 2021-05-18 Астразенека Аб Amino-triazolopyridine compounds and their use in treating cancer
CN110177791B (en) * 2016-12-20 2022-07-12 阿斯利康(瑞典)有限公司 Amino-triazolopyridine compounds and their use in the treatment of cancer
WO2018114999A1 (en) * 2016-12-20 2018-06-28 Astrazeneca Ab Amino-triazolopyridine compounds and their use in treating cancer
TWI776835B (en) * 2016-12-20 2022-09-11 瑞典商阿斯特捷利康公司 Amino-triazolopyridine compounds and their use in treating cancer
WO2018030881A3 (en) * 2017-01-03 2018-03-29 윤성문 Use of thyroid hormone for treatment of choroidal disease
JP7136793B2 (en) 2017-02-23 2022-09-13 ドメイネクス リミテッド 5-(pyrimidin-4-yl)-2-(pyrrolidin-1-yl)nicotinonitrile compounds as IKKE, TBK1 and/or SIK2 kinase inhibitors
JP2020508341A (en) * 2017-02-23 2020-03-19 ドメイネクス リミテッド 5- (Pyrimidin-4-yl) -2- (pyrrolidin-1-yl) nicotinonitrile Compounds as IKKE, TBK1 and / or SIK2 Kinase Inhibitors
US11878019B2 (en) 2017-02-28 2024-01-23 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US11285158B2 (en) 2017-02-28 2022-03-29 The General Hospital Corporation Uses of pyrimidopyrimidinones as SIK inhibitors
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
CN114127057A (en) * 2019-05-10 2022-03-01 德西费拉制药有限责任公司 Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
US11530206B2 (en) 2019-05-10 2022-12-20 Deciphera Pharmaceuticals, Llc Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
US11518758B2 (en) 2019-05-10 2022-12-06 Deciphera Pharmaceuticals, Llc Heteroarylaminopyrimidine amide autophagy inhibitors and methods of use thereof
US11590134B2 (en) 2019-06-17 2023-02-28 Deciphera Pharmaceuticals, Llc Aminopyrimidine amide autophagy inhibitors and methods of use thereof
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
CN113493471A (en) * 2020-04-03 2021-10-12 山东轩竹医药科技有限公司 Heteroaromatic kinase inhibitors
WO2022199561A1 (en) * 2021-03-23 2022-09-29 杭州阿诺生物医药科技有限公司 Hpk1 kinase inhibitor compound
WO2022251584A1 (en) * 2021-05-28 2022-12-01 St. Jude Children's Research Hospital, Inc. Methods and compositions for transduction of nk cells
CN115650956A (en) * 2022-10-11 2023-01-31 深圳市龙华区中心医院 Small molecule compound for targeted inhibition of activity of human and mouse RIPK3 protein kinases

Also Published As

Publication number Publication date
US20100056524A1 (en) 2010-03-04

Similar Documents

Publication Publication Date Title
WO2009122180A1 (en) Pyrimidine derivatives capable of inhibiting one or more kinases
US20210061803A1 (en) Inhibitors of cyclin-dependent kinase 7 (cdk7)
EP2403853B1 (en) Pyrrolopyrimidines used as kinase inhibitors
US20190292167A1 (en) Heteroaromatic compounds useful for the treatment of proliferative diseases
EP2118076B1 (en) Quinoxaline compounds and use thereof
US9428508B2 (en) 2,4-diamino-6,7-dihydro-5H-pyrrolo[2,3]pyrimidine derivatives as FAK/Pyk2 inhibitors
AU2006297089B2 (en) Diarylamine-containing compounds and compositions, and their use as modulators of c-kit receptors
US20110160185A9 (en) Pyrrolopyrimidine derivatives as jak3 inhibitors
TW202115065A (en) Kras mutant protein inhibitor
WO2015154022A1 (en) Inhibitors of cyclin-dependent kinase 7 (cdk7)
CA3050853A1 (en) Heterocyclic spiro compounds as magl inhibitors
AU2006327876A1 (en) Pyrimidine derivatives
KR20140026627A (en) Indazoles
AU2010224693A1 (en) Compounds
CN107454902B (en) Chromene derivatives as phosphoinositide 3-kinase inhibitors
EP3423445B1 (en) Novel compounds and pharmaceutical compositions thereof for the treatment of fibrosis
JP2007536365A (en) 3- or 4-monosubstituted phenol and thiophenol derivatives useful as H3 ligands
WO2005068468A2 (en) Heterocyclic protein kinase inhibitors and uses thereof
TW202344508A (en) Thiocarbamate derivatives as a2a inhibitors and methods for use in the treatment of cancers
CN114340634A (en) Heterocyclic compounds as kinase inhibitors
US20230227429A1 (en) Sulphonamide compounds
WO2023067546A1 (en) Novel bicyclic heteroaryl derivatives as sos1:kras proteinprotein interaction inhibitors
CN115151550A (en) Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP 1) modulators and uses thereof
WO2023109751A1 (en) Pyrimidine or pyridine derivative and medicinal use thereof
CN107743489B (en) 7- (morpholin-4-yl) pyrazolo [1,5-a ] pyrimidine derivatives for the treatment of immune, inflammatory or cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09726516

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09726516

Country of ref document: EP

Kind code of ref document: A1